WO2023163033A1 - NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF - Google Patents

NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF Download PDF

Info

Publication number
WO2023163033A1
WO2023163033A1 PCT/JP2023/006432 JP2023006432W WO2023163033A1 WO 2023163033 A1 WO2023163033 A1 WO 2023163033A1 JP 2023006432 W JP2023006432 W JP 2023006432W WO 2023163033 A1 WO2023163033 A1 WO 2023163033A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
synuclein
group
brain
ethyl acetate
Prior art date
Application number
PCT/JP2023/006432
Other languages
French (fr)
Japanese (ja)
Inventor
真人 樋口
麻衣子 小野
明栄 張
裕平 高堂
究 松岡
広 水間
武志 山本
健志 若林
俊行 大房
Original Assignee
国立研究開発法人量子科学技術研究開発機構
エーザイ・アール・アンド・ディー・マネジメント株式会社
小野薬品工業株式会社
武田薬品工業株式会社
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 国立研究開発法人量子科学技術研究開発機構, エーザイ・アール・アンド・ディー・マネジメント株式会社, 小野薬品工業株式会社, 武田薬品工業株式会社 filed Critical 国立研究開発法人量子科学技術研究開発機構
Publication of WO2023163033A1 publication Critical patent/WO2023163033A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/06Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms

Definitions

  • the present invention relates to a novel compound, an ⁇ -synuclein aggregate binding agent and uses thereof, and in particular, a novel compound, an ⁇ -synuclein aggregate binding agent containing the novel compound, a composition for optical imaging of ⁇ -synuclein aggregate, ⁇
  • the present invention relates to a composition for radioimaging of brain ⁇ -synuclein aggregates, an optical imaging method of brain ⁇ -synuclein aggregates, a radioimaging method of brain ⁇ -synuclein aggregates, and intermediates for synthesizing novel compounds.
  • ⁇ -Synuclein is a protein localized in synapses of neurons. Aggregates of ⁇ -synuclein (hereinafter referred to as ⁇ -synuclein aggregates) form the core pathology of Parkinson's disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), and have a close causal relationship with neurodegeneration. are considered to have Definitive diagnosis of such diseases is made by pathological analysis of the brain at autopsy using the presence of ⁇ -synuclein aggregates (also referred to herein as " ⁇ -synuclein lesions”) as an indicator, and thus the diagnosis cannot be confirmed before death.
  • ⁇ -synuclein aggregates also referred to herein as " ⁇ -synuclein lesions
  • ⁇ -synuclein aggregates can be visualized in the living brain, it will be possible to obtain information close to definitive diagnosis of these diseases at an early stage.
  • ⁇ -synuclein aggregates can be visualized in the living brains of disease model animals, it will be useful for efficacy evaluation of therapeutic or preventive drug candidate substances that target ⁇ -synuclein aggregates by imaging over time.
  • [ 11 C]BF-227 is a PET (positron emission tomography) probe that has been shown to bind to ⁇ -synuclein in the living brain (Non-Patent Documents 1 and 2).
  • [ 11 C]BF-227 does not have sufficient binding affinity to ⁇ -synuclein aggregates, and among the above diseases, ⁇ -synuclein lesions can only be detected in some MSA patients.
  • [ 11 C]BF-227 also has problems of non-specific accumulation in the brain and low binding selectivity to ⁇ -synuclein aggregates because it binds to amyloid ⁇ aggregates.
  • Patent Document 1 a compound for imaging tau protein accumulated in the brain (see Patent Document 1). Since the compound described in Patent Document 1 can image tau protein accumulated in the brain, the technique of Patent Document 1 is useful for the treatment and prevention of diseases caused by the accumulation of tau protein, such as Alzheimer's disease (AD). However, Patent Document 1 does not describe binding to ⁇ -synuclein aggregates.
  • the present invention has been made in view of the above circumstances, and provides an ⁇ -synuclein aggregate-binding agent with high binding selectivity to ⁇ -synuclein aggregates, an imaging method using this ⁇ -synuclein aggregate-binding agent, and ⁇ -synuclein It is an object of the present invention to provide novel compounds that can be used as aggregate binders.
  • the present inventors have found that a compound having a specific structure has high binding selectivity to ⁇ -synuclein aggregates, and after further investigation, have completed the present invention. More specifically, the present invention provides the following.
  • One aspect of the present invention is a compound represented by formula (I), (II) or (III) below, or a pharmaceutically acceptable salt or solvate thereof.
  • an ⁇ -synuclein aggregate-binding agent with high binding selectivity to ⁇ -synuclein aggregates can be provided.
  • FIG. 4 shows fluorescence microscope measurement results of DLB patient brains and AD patient brains.
  • FIG. 10 is a diagram showing quantitative results of fluorescence intensity of a lesion area and a lesion-free area;
  • FIG. 3 shows fluorescence microscope measurement results of ⁇ -synuclein fiber-inoculated mice.
  • FIG. 2 shows the results of two-photon laser scanning fluorescence microscopy of ⁇ -synuclein fiber-inoculated mice.
  • a and/or B intends at least one of A and B.
  • pharmaceutically acceptable salt refers to salts that are not harmful to mammals, especially humans.
  • Pharmaceutically acceptable salts can be formed with non-toxic acids or bases, including inorganic acids or bases, or organic acids or bases.
  • examples of pharmaceutically acceptable salts include metal salts formed from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or lysine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, and organic salts formed from diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and the like.
  • Pharmaceutically acceptable salts also include acid addition salts and base addition salts.
  • pharmaceutically acceptable carrier means a pharmaceutically acceptable material, composition, or vehicle such as a saline solution, liquid or solid filler, diluent, solvent, or encapsulating material.
  • pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. , dextrose, and Lactated Ringer's Injection.
  • an effective amount refers to the amount of a compound or composition that can achieve the desired effect.
  • an effective amount refers to that amount of a compound or composition that allows for optical or radioimaging of substances that accumulate in the brain, such as alpha-synuclein aggregates.
  • solvate means a solvate formed by the association of one or more solvent molecules with the compound.
  • Solvates include, for example, mono-, di-, tri-, and tetra-solvates. Solvates also include hydrates.
  • hydrate means a compound or salt thereof further containing a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces. Hydrates include, for example, monohydrates, dihydrates, trihydrates, tetrahydrates, and the like.
  • treatment means reducing or ameliorating the progression, severity and/or duration of a disease or condition.
  • prevention means reducing the risk of acquiring or developing a given disease or condition, or reducing or inhibiting the recurrence, onset, or progression of one or more symptoms of a given disease or condition. .
  • binding means the binding strength of a compound to a specific protein aggregate.
  • binding selectivity means that there is a difference in the binding properties of a compound when comparing the binding properties of a compound to a specific protein aggregate and the binding properties of other protein aggregates (specific protein aggregates). binding to aggregates is higher or lower than binding to other protein aggregates). “High binding selectivity” means that the difference in the above binding properties is large. For example, when a compound "has high selectivity for binding to ⁇ -synuclein aggregates", it means that there is a large difference between the binding ability of the compound to ⁇ -synuclein aggregates and the binding ability to other protein aggregates. It shows that the aggregates have higher binding properties.
  • the present invention provides (E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazin-2-yl)buta- 1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol, (E)-1-fluoro-3- represented by the following structural formula (II) ((2-(4-(6-methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol , or (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-ene-3-) represented by the following structural formula (III) In-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propan-2-ol, a pharmaceutically acceptable salt thereof, or
  • compound (I), compound (II) and compound (III) are compounds wherein one or more atoms are radioactive isotopes of said atoms, salts thereof, or solvates thereof.
  • Radioisotopes are selected from the group consisting of 15 O, 13 N, 11 C, and 18 F, but are not particularly limited.
  • the radioisotope is 11 C or 18 F. Considering that 11 C has a half-life of about 20 minutes and that of 18 F has a half-life of about 110 minutes, the 18 F-labeled compound is considered to have a higher commercial utility value. Conceivable. Therefore, most preferably the radioisotope is 18F .
  • a methylamino group bonded to a pyrazine ring or a pyridine ring, and a 3-fluoro-2-hydroxypropylamino group (-NH-CH 2 -CH(OH)-CH 2 bonded to a benzothiazole ring)
  • At least one of F) or a 3-fluoro-2-hydroxypropoxy group (—O—CH 2 —CH(OH)—CH 2 F) bonded to the thiazolopyridine ring is a group containing a radioactive isotope.
  • a 3-fluoro-2-hydroxypropylamino group or a 3-fluoro-2-hydroxypropoxy group is a group containing a radioisotope.
  • the fluorine atom in the 3-fluoro-2-hydroxypropylamino group or 3-fluoro-2-hydroxypropoxy group is a radioactive isotope.
  • the compound (I) containing a radioisotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazine-2 -yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol.
  • the compound (II) containing a radioisotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(6-methylamino)pyridine-3 -yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol.
  • the compound (III) containing a radioactive isotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazine- 2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propan-2-ol.
  • the compound represented by the following formula (IV) is an intermediate compound for the production of compound (I) and compound (II).
  • the compound represented by the following formula (V) is a production intermediate compound of compound (III).
  • the compound represented by the following formula (VI) is an intermediate compound for the production of (I) and compound (II).
  • these production intermediate compounds are also referred to as “intermediate compounds” or simply "intermediates”.
  • R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group (tosyl group) or a methanesulfonyl group.
  • R 2 is a tetrahydro-2H-pyran-2-yl group or a methoxymethyl group.
  • R 4 is a hydrogen atom, a tert-butoxycarbonyl (Boc) group or a 2,4-dimethoxybenzyl group.
  • R 3 is a hydrogen atom, tert-butoxycarbonyl group or 2,4-dimethoxybenzyl group.
  • X is a nitrogen atom or an unsubstituted carbon atom.
  • unsubstituted carbon atom refers to CH.
  • R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group
  • R 4' is a hydrogen atom or a tert-butoxycarbonyl group
  • X is a nitrogen atom (N) or an unsubstituted carbon atom (CH).
  • intermediate compounds can be used to synthesize compound (I) and compound (II) or synthesize compound (III), further synthesize radioisotope-labeled compound (I) and compound (II), or radioisotope-labeled It is preferably used for synthesizing compound (III). Also, these intermediate compounds may be salts.
  • compound (I), compound (II) or compound (III) contains various isomers such as stereoisomers (including optical isomers and rotational isomers), tautomers, or polar isomers, respectively are also included in compound (I), compound (II) or compound (III). These isomers can be obtained as single products by known synthetic techniques and separation techniques. Compound (IV), compound (V) and compound (VI) may also include various isomers.
  • Compounds (I) to (VI) may also be crystals produced by known crystallization methods.
  • Compounds (I) to (VI) can be produced according to the production method shown below. Further, if desired, deprotection reaction, amidation reaction, urea reaction, alkylation reaction, Mitsunobu reaction, oxidation reaction, reduction reaction, halogenation reaction, coupling reaction, nucleophilic addition reaction with carbanion, Grignard reaction, dehydration.
  • Compounds (I) to (VI) can be produced by performing each reaction alone or in combination of two or more thereof.
  • reaction conditions such as solvents, reagents and temperatures in each reaction may be appropriately set based on the common technical knowledge of those skilled in the art. Protection and deprotection reactions of functional groups are carried out according to known reaction methods and methods described in Reference Examples or Examples, and conventional protecting groups are used.
  • compound (1), compound (2), etc. [1. Method for producing compound (I), compound (II), and its intermediate compound (IV)] (Manufacturing method A)
  • compound (I) and compound (II) can be produced by the following method shown in Production Scheme 1.
  • TBS represents a tert-butyldimethylsilyl group.
  • Compound (3) can be produced by an alkylation reaction of compound (1) through an oxirane ring-opening reaction of compound (2).
  • Compound (4) can be produced by hydroxyl-protecting compound (3) with tert-butyldimethylsilyl chloride.
  • Compound (5) can be produced by amino group-protecting reaction of compound (4) with di-tert-butyl dicarbonate.
  • Compound (6) can be produced by formylating compound (5) with N,N-dimethylformamide or the like.
  • Compound (7) can be produced by a vinyl bromination reaction of compound (6) using (bromomethyl)triphenylphosphonium bromide.
  • Compound (9) can be produced by the Sonogashira reaction between compound (7) and compound (8) described below.
  • Compound (I) and compound (II) can be produced by a deprotection reaction of compound (9).
  • Compound (8) used in Production Method A can be produced from Compound (10) by the following method shown in Production Scheme 2.
  • Hal represents a halogen atom (eg, bromine atom or iodine atom), and TMS represents a trimethylsilyl group.
  • Compound (11) can be produced by amino group-protecting (di-Boc) reaction of compound (10) with di-tert-butyl dicarbonate.
  • Compound (12) can be produced by a deprotection (mono-Boc) reaction of one Boc group in compound (11) with a base such as potassium carbonate in a solvent such as methanol.
  • Compound (13) can be produced by a methylation reaction of compound (12) with iodomethane or the like.
  • Compound (15) can be produced by a Sonogashira reaction between compound (13) and ethynyltrimethylsilane (14).
  • Compound (8) can be produced by a deprotection reaction of the TMS group of compound (15).
  • compound (I) and compound (II) can also be produced by the following method shown in Production Scheme 3.
  • Compound (17) can be produced by formylating compound (16) with N,N-dimethylformamide or the like.
  • Compound (18) can be produced by a vinyl bromination reaction of compound (17) using (bromomethyl)triphenylphosphonium bromide.
  • Compound (19) can be produced by the Sonogashira reaction of compound (18) and compound (8).
  • Compound (20) can be produced by a deprotection reaction of compound (19).
  • Compound (I) and compound (II) can be produced by an oxirane ring-opening reaction of compound (2) and an alkylation reaction of compound (20).
  • compound (2) having a fluorine atom of 18 F that is, [ 18 F]2-(fluoromethyl)oxirane (CAS[ 123436-06-6])
  • compounds (I) and compounds (II) in which the fluorine atom in the 3-fluoro-2-hydroxypropylamino group is a radioactive isotope can be produced.
  • [ 18 F]2-(fluoromethyl)oxirane can be synthesized by nucleophilic substitution of glycidyl tosylate with [ 18 F]fluoride ion and purification by distillation. An example of the synthesis of [ 18 F]2-(fluoromethyl)oxirane is shown below.
  • compound (IV) (compound (IV-i)) in which R 3 and R 4 are Boc groups can be produced by the following method shown in Production Scheme 4.
  • R 5 represents a halogen atom (eg, bromine atom or iodine atom) or a leaving group such as a 4-nitrobenzenesulfonyloxy group.
  • Compound (IV-i) can be produced by an alkylation reaction of compound (19) with compound (21), which will be described later, or the like.
  • Compound (21a) in which R 5 is a halogen atom and R 1 is a tosyl group or a 4-nitrobenzenesulfonyl group in Compound (21) used in Production Method C can be prepared by the following method shown in Production Scheme 5 to Compound (22 ).
  • R 1a represents a tosyl group or a 4-nitrobenzenesulfonyl group
  • Hal represents a halogen atom (eg, iodine atom).
  • Compound (23) can be produced by a sulfonylation reaction of compound (22).
  • Compound (21a) can be produced by a hydroxyl group-protecting reaction of compound (23) with a tetrahydropyranyl (THP) group, a methoxymethyl (MOM) group, or the like.
  • THP tetrahydropyranyl
  • MOM methoxymethyl
  • Compound (33) can be produced by a bromination reaction of compound (32).
  • Compound (34) can be produced by reacting compound (33) with triethyl phosphite.
  • Compound (35) can be produced by an alkoxylation reaction of compound (34) with 3,4-dimethoxybenzyl alcohol.
  • Compound (37) can be produced by Horner-Wadsworth-Emmons reaction (HWE reaction) between compound (35) and compound (36) described below.
  • Compound (38) can be produced by a deprotection reaction of compound (37).
  • Compound (III) can be produced by an oxirane ring-opening reaction of compound (39) and an alkylation reaction to compound (38).
  • Compound (36) used in Production Method D can be produced from Compound (40) by the following method shown in Production Scheme 7.
  • compound (39) in which the fluorine atom is 18 F that is, [ 18 F]2-(fluoromethyl)oxirane is used.
  • compound (III) in which the fluorine atom in the 3-fluoro-2-hydroxypropoxy group is a radioactive isotope can be prepared.
  • Compound (41) can be produced by amino group-protecting (di-Boc) reaction of compound (40) with di-tert-butyl dicarbonate.
  • Compound (42) can be produced by a deprotection (mono-Boc) reaction of one Boc group in compound (41) with a base such as potassium carbonate in a solvent such as methanol.
  • Compound (43) can be produced by a methylation reaction of compound (42) with iodomethane or the like.
  • Compound (45) can be produced by a Sonogashira reaction between compound (43) and propargyl alcohol (44).
  • Compound (36) can be produced by oxidation reaction of compound (45).
  • compound (V) (compound (Vi)) in which R 4 is H can be produced by the following method shown in Production Scheme 8.
  • TBS represents a tert-butyldimethylsilyl group.
  • Compound (47) can be produced by an alkylation reaction such as Mitsunobu reaction between compound (38) and compound (46) described below.
  • Compound (48) can be produced by a deprotection reaction of compound (47).
  • Compound (Vi) can be produced by protecting compound (48).
  • compound (46a) in which R 1 is a p-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group can be produced from compound (49) by the following method shown in production scheme 9. can be done.
  • R 1b represents a p-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group.
  • Compound (50) can be produced by hydroxyl group protection reaction of compound (49).
  • Compound (46a) can be produced by a debenzylation reaction of compound (50).
  • compound (Vi) can be produced by the following method shown in Production Scheme 10.
  • R 5 represents a leaving group such as a halogen atom (eg, bromine atom or iodine atom), paratoluenesulfonyloxy group or 4-nitrobenzenesulfonyloxy group.
  • a halogen atom eg, bromine atom or iodine atom
  • paratoluenesulfonyloxy group e.g, paratoluenesulfonyloxy group or 4-nitrobenzenesulfonyloxy group.
  • Compound (Vi) can be produced by, for example, alkylating compound (38) with compound (51) described below.
  • the compound (51) used in production method F in which R 5 is a halogen atom and R 1 is a para-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group is the compound described in [2.
  • Compound (21a) in Production Scheme 5 of Compound (I), Compound (II), and Compound (IV), which is an intermediate thereof] can be produced by the method shown in Production Scheme 5.
  • Compound (III) can be produced by a deprotection reaction after a fluorination reaction of production intermediate compound (V) with potassium fluoride or the like. Further, by using [ 18 F]potassium fluoride or the like, compound (III), which is a radioactive isotope, can be produced.
  • Compound (53) can be produced by subjecting compound (52) to a tosylation reaction.
  • Compound (54) can be produced by oxidation reaction of the hydroxyl group of compound (53).
  • Compound (55) can be produced by an imine formation reaction between compound (20) and compound (54).
  • Compound (56) can be produced by a reduction reaction of the imino group of compound (55).
  • Compound (57) can be produced by a deprotection reaction of compound (56).
  • Compound (58) can be produced by a reduction reaction of the carbonyl group of compound (57).
  • Compound (VI-i) can be produced by a hemiaminal etherification reaction between the amino group and hydroxyl group of compound (58).
  • the invention provides alpha-synuclein aggregate binding agents.
  • the ⁇ -synuclein aggregate binding agent (hereinafter also referred to as a binding agent) of this embodiment is compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof. contains.
  • Compound (I), compound (II) and compound (III) have higher binding selectivity to ⁇ -synuclein aggregates than to aggregates of tau protein and amyloid ⁇ .
  • pharmaceutically acceptable salts of Compound (I), Compound (II) and Compound (III), and solvates of Compound (I), Compound (II) and Compound (III) also include tau protein. and more selective binding to ⁇ -synuclein aggregates than to aggregates of amyloid ⁇ .
  • compound (I), compound (II) and compound (III) emit fluorescence.
  • one or more atoms can be a radioactive isotope of that atom. Therefore, the binding agents of this embodiment can be used as molecular probes for optical or radioimaging of ⁇ -synuclein aggregates accumulated in the brain.
  • the ⁇ -synuclein aggregate binding agent can contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
  • the content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the ⁇ -synuclein aggregate-binding agent is There are no particular restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors.
  • the content of pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the alpha-synuclein aggregate binding agent.
  • the ⁇ -synuclein aggregate binding agent can be administered with compound (I), compound (II) or compound (III), for example, in an amount of 5 ng/kg to 5 mg/kg of the compound per body weight (kg) of the subject.
  • the lower limit of the amount of the compound is 5 ng/kg or more, 0.01 mg/kg or more, 0.05 mg/kg or more, or 0.1 mg/kg or more.
  • the upper limit of the amount of the compound is 5 mg/kg or less, 3 mg/kg or less, 1 mg/kg or less, or 20 ⁇ g/kg or less.
  • composition for optical imaging of ⁇ -synuclein aggregates contains the above-described binder according to the present embodiment.
  • Such optical imaging includes in vitro, ex vivo, and in vivo imaging.
  • Optical imaging includes, for example, fluorescence microscopy, multiphoton imaging, two-photon imaging, and near-infrared fluorescence imaging.
  • the optical imaging composition can contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
  • the content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the optical imaging composition is There are no restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors.
  • the content of the pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the optical imaging composition.
  • composition for optical imaging contains compound (I), compound (II) or compound (III), for example, the compound amount (mg) per subject body weight (kg) is 0.01 mg/kg to 5 mg/kg, It is prepared so that it can be administered in an amount of preferably 0.05 mg/kg to 3 mg/kg, more preferably 0.1 mg/kg to 1 mg/kg.
  • composition for radiation imaging of ⁇ -synuclein aggregates provides a composition for radioimaging alpha-synuclein aggregates.
  • the composition for radiation imaging of ⁇ -synuclein aggregates of the present embodiment (hereinafter also simply referred to as "composition for radiation imaging") is compound (I), compound (II) and compound (III), or Includes binders according to this embodiment described above that contain compounds in which a further atom is a radioactive isotope of that atom.
  • Such radioimaging includes in vitro, ex vivo, and in vivo imaging.
  • Radiation imaging includes, for example, positron emission tomography (PET), single photon emission computed tomography (SPECT), and autoradiography.
  • PET positron emission tomography
  • SPECT single photon emission computed tomography
  • autoradiography includes, for example, positron emission tomography (PET), single photon emission computed tomography (SPECT), and autoradiography.
  • the radioimaging composition can contain a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
  • the content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the radioimaging composition is particularly There are no restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors.
  • the content of the pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the radioimaging composition.
  • the radiation imaging composition contains compound (I), compound (II) or compound (III), for example, in an amount of the compound per body weight (kg) of the subject of 5 ng/kg to 5 mg/kg, preferably 5 ng/kg. It is formulated to be dosed at ⁇ 20 ⁇ g/kg.
  • the present invention provides diagnostics for diseases associated with ⁇ -synuclein aggregates, or companion diagnostics for the treatment or prevention of such diseases.
  • the diagnostic agent for diseases associated with ⁇ -synuclein aggregates of the present embodiment, or the companion diagnostic agent for treating or preventing the disease (hereinafter also referred to as the companion diagnostic agent) is the binding agent according to the present embodiment described above.
  • a companion diagnostic for treatment is a diagnostic that, if found to be diseased, is used to determine whether treatment is likely.
  • companion diagnostics for prevention are diagnostics for predicting future onset or judging whether prevention can be expected to suppress onset when it turns out to be a prodromal state of a disease. be.
  • diagnostic agent Using the diagnostic agent according to the present embodiment, data on the amount and/or distribution of brain ⁇ -synuclein aggregates obtained from a subject are compared with the previously obtained disease and the amount and/or distribution of ⁇ -synuclein aggregates. By checking the correlation, it is possible to diagnose the target disease (specifically, the presence or absence of the disease, the severity, the possibility of seizure, etc.).
  • ⁇ -synuclein aggregates obtained from a subject can be obtained by comparing the previously obtained disease with the amount and/or distribution of brain ⁇ -synuclein aggregates. By checking the correlation, the disease state of the subject can be grasped. Therefore, based on this, disease prophylaxis/treatment regimens (types of prophylactic or therapeutic agents to be administered, combinations, doses, uses, etc.) can be formulated.
  • One embodiment of the present invention is a medicament for the treatment or prevention of diseases associated with ⁇ -synuclein aggregates, which is administered based on data on the amount and/or distribution of brain ⁇ -synuclein aggregates obtained by companion diagnostics. It also relates to medicaments administered on a schedule.
  • a diagnostic kit for a disease associated with a substance that accumulates in the brain of the present invention includes the above-described binding agent according to this embodiment.
  • Substances that accumulate in the brain include at least ⁇ -synuclein aggregates, and also include tau protein or amyloid ⁇ aggregates.
  • Diseases associated with substances that accumulate in the brain include at least diseases associated with ⁇ -synuclein aggregates, such as Parkinson's disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). is mentioned.
  • Other diseases associated with substances that accumulate in the brain include Alzheimer's disease (AD) and frontotemporal lobar degeneration, which are diseases associated with aggregates of tau protein or amyloid ⁇ .
  • AD Alzheimer's disease
  • frontotemporal lobar degeneration which are diseases associated with aggregates of tau protein or amyloid ⁇ .
  • the binding agent of the present embodiment has higher binding selectivity to ⁇ -synuclein aggregates than to aggregates of tau protein and amyloid ⁇ .
  • the results of studies by the present inventors revealed that, for example, the compounds described in Patent Document 1 have higher binding properties to tau protein aggregates than to ⁇ -synuclein aggregates.
  • the diagnostic kit comprises compound (I), compound (II) or compound (III) with high binding selectivity to ⁇ -synuclein aggregates or a pharmaceutically acceptable salt or solvate thereof and other compounds with high binding selectivity to tau protein aggregates (for example, compounds described in Patent Document 1).
  • the former detection result amount and/or distribution of detected light or radiation
  • the latter detection result it is possible to determine which substance is present in each region imaged. can be identified. Specifically, ⁇ -synuclein aggregates and tau protein aggregates can be classified, and the abundance of each can be quantified.
  • diseases associated with ⁇ -synuclein aggregates and/or diseases associated with tau protein can be diagnosed with high accuracy.
  • the binding agent of the present embodiment and a substance with high binding selectivity to tau protein aggregates are specified in advance. Then, by measuring the amount ratio of light or radiation in each area of the brain after administration of the former and the latter in the subject, ⁇ -synuclein aggregates, ⁇ -synuclein aggregates, It is possible to classify which tau protein aggregates are present and quantify the amount of each.
  • the diagnostic kit of the present embodiment can be further combined with an amyloid ⁇ imaging agent to form a diagnostic kit.
  • a diagnostic kit further combined with an amyloid ⁇ imaging agent can specify which substance is present in each region imaged. Specifically, ⁇ -synuclein aggregates, tau protein aggregates, and amyloid ⁇ aggregates can be classified, and the abundance of each can be quantified. Therefore, diseases associated with ⁇ -synuclein aggregates, diseases associated with tau protein, and/or diseases associated with amyloid ⁇ can be diagnosed with high accuracy.
  • One embodiment of the present invention is a medicament for treating or preventing diseases associated with ⁇ -synuclein aggregates, wherein the amount of brain accumulated substances containing ⁇ -synuclein aggregates obtained from the diagnostic kit according to this embodiment is and/or a medicament administered in a dosing regimen based on distribution data.
  • the invention provides an optical imaging method.
  • the brain of a living subject to whom the binding agent of the present embodiment has been administered is irradiated with light of a first wavelength from outside the brain, the first detecting light of a second wavelength different from the wavelength of the.
  • the binding agent transferred to the living brain binds to ⁇ -synuclein aggregates present in the living brain.
  • Light of a first wavelength that excites the binding agent is irradiated extracerebrally into the living brain of a subject administered the binding agent, and light of a second wavelength (e.g., fluorescence) emitted from the binding agent in the brain.
  • a second wavelength e.g., fluorescence
  • Subjects include mammals. Mammals include, for example, humans, rats, mice, rabbits, guinea pigs, hamsters, monkeys, dogs, ferrets, minipigs, and the like. Preferably, the mammal is human.
  • the administration method is not particularly limited, but includes, for example, oral administration and parenteral administration such as intravenous administration or intraperitoneal administration. Intravenous administration or intraperitoneal administration is preferred. Most preferred is intravenous administration.
  • the dosage is such that the amount (mg) of compound (I), compound (II) or compound (III) per body weight (kg) of the subject is 0.01 mg/kg to 5 mg/kg, 0.05 mg/kg to 3 mg. /kg, or 0.1 mg/kg to 1 mg/kg, most preferably 0.1 mg/kg to 1 mg/kg.
  • the invention provides a radiation imaging method.
  • the radioimaging method of this embodiment comprises compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a Detecting radiation emitted from the living brain of a subject administered a binding agent according to the present embodiments, including the solvate.
  • the binding agent transferred to the living brain binds to ⁇ -synuclein aggregates present in the living brain.
  • Radiation imaging of ⁇ -synuclein aggregates can be performed by detecting radiation emitted from the binding agent in the brain.
  • Subjects include mammals. Mammals include, for example, humans, rats, mice, rabbits, guinea pigs, hamsters, monkeys, dogs, ferrets, minipigs, and the like. Preferably, the mammal is human.
  • the administration method is not particularly limited, but includes, for example, oral administration and parenteral administration such as intravenous administration or intraperitoneal administration. Intravenous administration or intraperitoneal administration is preferred. Most preferred is intravenous administration.
  • the dosage is preferably 5 ng/kg to 5 mg/kg of compound (I), compound (II) or compound (III) per body weight (kg) of the subject, and 5 ng to 20 ⁇ g/kg. kg is more preferred.
  • the amount of radioactivity administered is preferably 37 MBq to 7.4 GBq, more preferably 370 MBq to 3,700 MBq per individual.
  • the present invention provides a method of screening for therapeutic or preventive agents for diseases associated with brain ⁇ -synuclein aggregates.
  • the screening method for therapeutic or preventive drugs for diseases associated with brain ⁇ -synuclein aggregates of the present embodiment comprises optical imaging according to the present embodiment before and after administration of a candidate substance to a subject. selecting candidate substances based on differences in the amount and/or distribution of light or radiation detected by the method or radiation imaging method.
  • ⁇ -synuclein aggregates are at least the same as the diseases associated with ⁇ -synuclein aggregates described in the above [Diagnostic kit for diseases associated with substances that accumulate in the brain].
  • the candidate substance is associated with the disease or condition.
  • amount (intensity) of light or radiation such as fluorescence of the binding agent
  • Said candidate compound may be useful as a therapeutic compound for said disease or condition.
  • data on the amount (intensity) and/or distribution of light or radiation such as fluorescence of a binding agent before and after administration of a candidate substance obtained from a subject to whom the candidate substance has been administered may be collected from a previously acquired mammal not administered the candidate substance.
  • the increase and/or change in distribution of light or radiation such as fluorescence of the binding agent before and after the onset of disease involving brain ⁇ -synuclein aggregates in animals is compared.
  • a candidate substance may be useful as a preventive compound for the disease or condition if it exhibits values close to those of normal mammals as before administration.
  • the change in the distribution of light such as fluorescence or radiation seen after the onset of the disease is suppressed by administration of the candidate substance, and/or the change in the distribution of light such as fluorescence or radiation is suppressed by the candidate substance.
  • a candidate substance may also be useful as a compound for prophylaxis of the disease or condition if, after administration of the drug, the distribution in a normal mammal is as close to that as before administration.
  • the invention provides a method of quantifying or determining the accumulation of brain ⁇ -synuclein aggregates.
  • the method of quantifying or determining the accumulation of brain ⁇ -synuclein aggregates of this embodiment includes compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • This method is a method for quantifying or determining the accumulation of brain ⁇ -synuclein aggregates by optical imaging.
  • the subject and administration method are the same as those described in [Optical Imaging Method] above.
  • This method is a method for quantifying or determining the accumulation of ⁇ -synuclein aggregates in the brain by radioimaging.
  • the subject and administration method are the same as those described in [Radiation Imaging Method] above.
  • the invention provides a method for determining the classification and accumulation of substances that accumulate in the brain.
  • the method for determining the classification and accumulation of substances that accumulate in the brain of the present invention includes compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • a substance with high binding selectivity to tau protein aggregates (for example, the compound described in Patent Document 1) is administered to the living brain of the subject at a different time from the first step.
  • This method uses optical imaging to determine the classification and accumulation of substances that accumulate in the brain.
  • the subject and administration method are the same as those described in [Optical Imaging Method] above.
  • Accumulating in the brain based on the amount and/or distribution data of radiation detected in the first step and the amount and/or distribution data of radiation detected in the second step Determine the classification and accumulation of substances.
  • This method is a method of determining the classification and accumulation of substances that accumulate in the brain by radiation imaging.
  • the subject and administration method are the same as those described in [Radiation Imaging Method] above.
  • a first step of detecting radiation emitted from the brain due to administration of a binding agent with high binding selectivity to ⁇ -synuclein aggregates and a substance with high binding selectivity to tau protein aggregates in the same subject Since it includes both a second step of detecting radiation emitted from the brain resulting from the administration of the detection result of the first step (amount and / or distribution of radiation detected) and the detection of the second step By comparing the results, it is possible to determine whether the substances accumulated in the brain are ⁇ -synuclein aggregates and/or tau protein aggregates, and the accumulation of each aggregate.
  • One aspect of the present invention is a compound represented by formula (I), (II) or (III) below, or a pharmaceutically acceptable salt or solvate thereof.
  • One aspect of the present invention is a pharmaceutically acceptable or a solvate thereof.
  • One aspect of the present invention is an ⁇ -synuclein aggregate binding agent containing the compound, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  • One aspect of the present invention is a composition for optical imaging of ⁇ -synuclein aggregates, containing the ⁇ -synuclein aggregate-binding agent.
  • One aspect of the present invention is a composition for radioimaging of ⁇ -synuclein aggregates, which contains the ⁇ -synuclein aggregate-binding agent.
  • the living brain of a subject to whom the ⁇ -synuclein aggregate-binding agent is administered is irradiated with light of a first wavelength from outside the brain
  • the first wavelength emitted from the brain is A method of optical imaging brain ⁇ -synuclein aggregates comprising detecting light of a different second wavelength.
  • One aspect of the present invention is a radioimaging method for intracerebral ⁇ -synuclein aggregates, comprising the step of detecting radiation emitted from the living brain of a subject administered with the ⁇ -synuclein aggregate-binding agent.
  • R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group
  • R 2 is a tetrahydro-2H-pyran-2-yl group or a methoxymethyl group
  • R 4 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group
  • X is a nitrogen atom (N) or an unsubstituted carbon atom (CH);
  • R 3 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group.
  • R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group
  • R 4' is a hydrogen atom or a tert-butoxycarbonyl group
  • X is a nitrogen atom (N) or an unsubstituted carbon atom (CH).
  • ACD/SpecManager (trade name) software or the like was used for 1 H NMR analysis, and the analytical values obtained were described. Peaks with very slow proton peaks, such as hydroxyl groups and amino groups, are sometimes not described.
  • Boc tert-butoxycarbonyl group
  • DMF N,N-dimethylformamide
  • DMSO dimethylsulfoxide
  • DMSO-d 6 heavy dimethylsulfoxide 1 H NMR: proton nuclear magnetic resonance M: molarity MeOH: methanol
  • TBS tert-butyldimethylsilyl group
  • TEA triethylamine
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • THP tetrahydropyranyl group
  • TMS trimethylsilyl group
  • Ts paratoluenesulfonyl group .
  • Reference Example T004 Preparation of tert-butyl methyl (5-((trimethylsilyl)ethynyl)pyrazin-2-yl)carbamate tert-butyl (5-iodopyrazin-2-yl)(methyl)carbamate (1.95 g) prepared in Reference Example T003, ethynyltrimethylsilane (1.62 ml) and dichlorobis(triphenylphosphine)palladium(II) (123 mg) ) was added to a mixture of THF (6 ml) and TEA (6 ml) at room temperature under nitrogen atmosphere.
  • Reference Example T005 Preparation of tert-butyl (5-ethynylpyrazin-2-yl) (methyl) carbamate tert-butyl methyl (5-((trimethylsilyl)ethynyl)pyrazin-2-yl)carbamate prepared in Reference Example T004 (1.98 g, 22% by weight of 1,4-bis(trimethylsilyl)buta-1,3-diyne To a 0° C. solution of THF (20 ml) was added tetra-n-butylammonium fluoride (1 M THF solution, 11.1 ml). The mixture was stirred at 0° C.
  • tert-butyl (2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)(2-formylbenzo[d ]thiazol-6-yl)carbamate (163 mg) in THF (5 ml) was added. The mixture was stirred at -78°C to room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate and water, neutralized with 5% aqueous citric acid solution, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure.
  • the mixture was stirred at room temperature for 2 hours, diluted with ethyl acetate, adjusted to pH about 4 with a 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate.
  • the extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (22 mg) as a yellow solid.
  • the mixture was stirred at room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate, adjusted to pH about 4 with 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate.
  • the extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (436 mg) as a yellow solid.
  • Cupric (I) (5.4 mg) was added at room temperature under a nitrogen atmosphere. The mixture was stirred at room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate, adjusted to pH about 4 with 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (140 mg) as a yellow solid.
  • Reference Example T018 Preparation of 3-iodo-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate
  • a THF (25 ml) solution of (2-hydroxy-3-iodopropyl 4-methylbenzenesulfonate (2.00 g) and 3,4-dihydro-2H-pyran (5.12 ml) prepared in Reference Example T017 para Pyridinium toluenesulfonate (1.41 g) was added at room temperature, the mixture was stirred at room temperature for 4 hours, para-toluenesulfonic acid monohydrate (534 mg) was added at room temperature, and the mixture was stirred at room temperature for 4 hours.
  • Reference Example T020 Preparation of 2,2-dimethoxy-3-oxopropyl 4-methylbenzenesulfonate 1,1,1-Triacetoxy-1,1-dihydro-1,1,1-triacetoxy-1,1-dihydro-1,1,1-triacetoxy-1,1-dihydro-1, 2-benzoiodoxol-3-(1H)-one (1.14 g) was added at room temperature. The mixture was stirred at room temperature for 2 hours, then cooled to 0°C, diluted with ethyl acetate and saturated aqueous sodium thiosulfate, neutralized with 5% aqueous sodium hydrogen carbonate and extracted with ethyl acetate.
  • the mixture was cooled to , diluted with ethyl acetate, neutralized with a 5% aqueous sodium hydrogencarbonate solution, and extracted with ethyl acetate.
  • the extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure.
  • the residual solid was suspended in ethyl acetate and collected by filtration, washed with ethyl acetate and dried to give the title compound (83 mg) as an orange solid.
  • the suspension was sonicated into a solution and then stirred at room temperature for 10 hours. After stirring, the mixture was cooled to 0° C., diluted with ethyl acetate and water, acidified to about pH 4 with a 5% aqueous citric acid solution, and then extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (50 mg) as a yellow solid.
  • Example 1 (E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d ] Production of thiazol-6-yl)amino)propan-2-ol (Compound (I), hereinafter also referred to as “SPAL-T-83”) tert-Butyl (E)-(2-(4-(5-((tert-butoxycarbonyl)(methyl)amino)pyrazin-2-yl)but-1-en-3-yn- prepared in Reference Example T011 1-yl)benzo[d]thiazol-6-yl)(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)carbamate (22 mg), TFA (0.900 ml) and water (0.100 ml) ) was stirred at room temperature for 4 hours, cooled to 0° C., diluted with eth
  • the extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure.
  • the residual solid was suspended in ethyl acetate, and the solid collected by filtration was washed with ethyl acetate and dried to give the title compound (11.5 mg) as an orange solid.
  • Example 2 (E)-1-fluoro-3-((2-(4-(6-methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d ] Production of thiazol-6-yl)amino)propan-2-ol (compound (II), hereinafter also referred to as “SPAL-T-95”)
  • a mixture of amine (40 mg), 2-(fluoromethyl)oxirane (CAS [503-09-3]) (0.047 ml) and methoxyethan-1-ol (6 ml) was heated at 150° C.
  • Example 3 (E)-3((tert-butoxycarbonyl)(2-(4-(5-((tert-butoxycarbonyl)(methyl)amino)pyrazin-2-yl)but-1-ene-3 -yn-1-yl)benzo[d]thiazol-6-yl)amino)-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate Preparation tert-Butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yne prepared in Reference Example T014 -1-yl)pyrazin-2-yl(methyl)carbamate (436 mg) in DMF (20 ml) at 0° C.
  • Example 4 (E)-3-(2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d]thiazole-6 -yl)oxazolidin-5-yl)methyl 4-methylbenzenesulfonate (E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) prepared in Reference Example T025 A mixture of benzo[d]thiazol-6-yl)amino)propyl 4-methylbenzenesulfonate (375mg) and paraformaldehyde (63mg) in acetonitrile (80ml) was stirred at 60°C for 0.5 hours, then paraformaldehyde (42mg).
  • Reference Example E002 Preparation of tert-butyl (5-iodopyrazin-2-yl) carbamate Potassium carbonate (4.76 g) was added to a methanol (50 ml) solution of di-tert-butyl 5-iodopyrazin-2-yl)-2-imidodicarbonate (4.8375 g) prepared in Reference Example E001 at room temperature. The mixture was stirred at room temperature for 6 hours and then at 70° C. for 1 hour. After cooling the reaction solution to room temperature, water and ethyl acetate were added, and the aqueous layer was extracted with ethyl acetate.
  • Reference Example E003 Preparation of tert-butyl (5-iodopyrazin-2-yl) (methyl) carbamate Cesium carbonate (7.11 g) and iodomethane (1.018 ml) were added to a solution of tert-butyl (5-iodopyrazin-2-yl)carbamate (3.5023 g) prepared in Reference Example E002 in DMF (55 ml) at 0°C. added with The mixture was allowed to stir at room temperature for 19 hours. Water and ethyl acetate were added to the reaction solution, and the aqueous layer was extracted with ethyl acetate.
  • Reference Example E004 Preparation of tert-butyl (5-(3-hydroxyprop-1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate
  • a suspension of tert-butyl (5-iodopyrazin-2-yl)(methyl)carbamate (3.2794 g) prepared in Reference Example E003 in triethylamine (9.55 ml), propargyl alcohol (1.129 ml), iodide Copper(I) (0.224 g) and bis(triphenylphosphine)palladium(II) chloride (0.137 g) were added and allowed to stir at room temperature for 90 minutes under a nitrogen atmosphere.
  • Reference Example E005 Preparation of tert-butyl methyl (5-(3-oxoprop-1-yn-1-yl)pyrazin-2-yl)carbamate A solution of oxalyl chloride (1.409 ml) in dichloromethane (40 ml) was slowly added with a solution of DMSO (1.747 ml) in dichloromethane (10 ml) at -78°C and stirred for 5 minutes. To the reaction solution was added tert-butyl (5-(3-hydroxyprop-1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate (2.1609 g) prepared in Reference Example E004 in dichloromethane (20 ml).
  • Reference Example E007 Preparation of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine 1
  • 5-chloro-2-methylthiazolo[5,4-b]pyridine (CAS: 109202-21-3,500 mg) prepared in Reference Example E006 in alpha, alpha, alpha-trifluorotoluene (18 ml), N- Bromosuccinimide (723 mg) and 2,2′-azobis(isobutyronitrile) (89 mg) were added and the mixture was allowed to stir at 80° C. for 2 hours.
  • 2,2′-Azobis(isobutyronitrile) (35.6 mg) was added to the mixture and the mixture was stirred at 80° C. for 2 hours.
  • Reference Example E008 Preparation of diethyl ((5-chlorothiazolo[5,4-b]pyridin-2-yl)methyl)phosphonate A mixture of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine (319 mg) prepared in Reference Example E007 and triethylphosphite (15 ml) was stirred at 100° C. for 2 hours. After the mixture was cooled to room temperature, it was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-100% ethyl acetate/n-heptane, 5% methanol/ethyl acetate) to give the title compound (305 mg).
  • Example 5 (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo [ Preparation 1 of 5,4-b]pyridin-5-yl)oxy)propan-2-ol (compound (III), hereinafter also referred to as “SPAL-E-17”) Crude (E)-2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4 prepared in Reference Example E011 2-(fluoromethyl)oxirane (CAS: 503-09-3, 12.8 ⁇ l) to a mixture of b]pyridin-5-ol (32.8 mg) and potassium carbonate (14.2 mg) in DMF (500 ⁇ l) was added at room temperature.
  • Example 6 (E)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4- Preparation of b]pyridin-5-yl)oxy)-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate (E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) prepared in Reference Example E013 To a solution of thiazolo[5,4-b]pyridin-5-yl)oxy)propyl 4-methylbenzenesulfonate (269 mg) in dichloromethane (5 ml) was added 3,4-dihydro-2H-pyran (0.091 ml) and p - Toluenesulfonic acid monohydrate (47.7 mg) was added at
  • Example 7 (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo [ Preparation of 5,4-b]pyridin-5-yl)oxy)propan-2-ol (Compound III) 2 4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8 .8.8] A solution of hexacosane (Kryptofix® 222) (18.2 mg) in acetonitrile (0.50 ml) was added at room temperature and the mixture was concentrated under reduced pressure at 40°C.
  • Acetonitrile (1.0 ml) was added to the resulting residue, and the mixture was concentrated at 40° C. under reduced pressure.
  • Acetonitrile (1.0 ml) was added again to the obtained residue, and the mixture was concentrated at 40° C. under reduced pressure.
  • Toluene (1.0 ml) was added to the resulting residue, and the mixture was concentrated under reduced pressure at 40°C.
  • Acetonitrile (1.0 ml) was added to the resulting residue, and the mixture was concentrated at 40° C. under reduced pressure. The resulting residue was dried at 100° C. for 10 minutes under reduced pressure.
  • arrowheads indicate fluorescence of compounds bound to ⁇ -synuclein aggregates in DLB patient brains
  • arrows indicate fluorescence of compounds bound to amyloid ⁇ aggregates in AD patient brains
  • asterisks indicate tau aggregates in AD patient brains. Fluorescence of bound compound is shown.
  • Analysis software (Image J) was used to quantify the fluorescence intensity of lesion areas and non-lesion forming areas (background). The results are shown in FIG. SPAL-T-83, SPAL-T-95 and SPAL-E-17 were used as test compounds.
  • SPAL-T-83, SPAL-T-95 and SPAL-E-17 were confirmed to bind to ⁇ -synuclein aggregates formed in DLB patient brains. . Also, the binding of SPAL-T-83, SPAL-T-95 and SPAL-E-17 to ⁇ -synuclein aggregates was significantly higher than to tau or amyloid ⁇ aggregates formed in AD patient brains. confirmed to be strong. Furthermore, the ratio of binding to alpha-synuclein aggregates to binding to tau or amyloid beta aggregates was confirmed to be higher than for C05-05.
  • Mouse ⁇ -synuclein is expressed as a recombinant protein in E. coli, extracted, and incubated in vitro to form insoluble aggregates.
  • this ⁇ -synuclein aggregate is inoculated into the striatum of mice, the ⁇ -synuclein aggregate propagates to surrounding areas via neural circuits, and several months later, ⁇ -synuclein lesions are observed in the cerebral neocortex ( Masuda-Suzukake et al. Acta Neuropathol Commun 2, 88, 2014; Shimozawa et al.
  • mouse ⁇ -synuclein was expressed in E. coli as a recombinant protein, extracted, and incubated in vitro to form insoluble ⁇ -synuclein aggregates.
  • ⁇ -synuclein aggregates were inoculated into the striatum of mice, ⁇ -synuclein aggregates propagated to the surrounding area via neural circuits, and several months later, ⁇ -synuclein lesions were observed in the cerebral neocortex. was done. Inoculation of this ⁇ -synuclein aggregate into the mouse striatum was performed by the following method.
  • the head of a 9-week-old C57/BL/6 male mouse anesthetized with 1.5% (v/v) isoflurane was dehaired, the scalp was disinfected with isodine, xylocaine was applied, and the scalp was incised. The skull was exposed. Then, a hole was drilled in the skull at a position of Bregma 0.05 mm and Lateral 2 mm, and 3 ⁇ l of ⁇ -synuclein fiber solution (mouse ⁇ -synuclein fiber 4 mg/ml in saline) was injected at a depth of 2 mm using a glass pipette. After that, the scalp was put back and sutured.
  • ⁇ -synuclein fiber solution mouse ⁇ -synuclein fiber 4 mg/ml in saline
  • the right side is anti-phosphorylated ⁇ -synuclein antibody staining
  • the left side is SPAL-T-83, SPAL-T-95 and SPAL-E-17 fluorescent staining.
  • arrowheads indicate lesions composed of phosphorylated ⁇ -synuclein in ⁇ -synuclein fiber-inoculated mouse brain and fluorescence of compounds bound to them.
  • the mouse was fixed under a two-photon laser fluorescence microscope, and 100 ⁇ l of physiological saline containing 5 mmol/l of sulforhodamine 101 was intraperitoneally administered, followed by in vivo two-photon fluorescence imaging at an excitation wavelength of 900 nm. went. After that, 50 ⁇ l of DMSO solution containing 0.1% of C05-05, SPAL-T-83, SPAL-T-95 or SPAL-E-17 was intraperitoneally administered, and 30 minutes after administration, biological two-photon irradiation was performed at an excitation wavelength of 900 nm. Fluorescence imaging was performed.
  • the detection wavelength for C05-05, SPAL-T-83, SPAL-T-95 and SPAL-E-17 was 500-550 nm, and the detection wavelength for sulforhodamine 101 was 573-648 nm.
  • the results are shown in FIG. In FIG. 4, arrows indicate blood vessels and arrowheads indicate fluorescence of compounds bound to ⁇ -synuclein lesions.
  • an ⁇ -synuclein aggregate-binding agent with high binding selectivity to ⁇ -synuclein aggregates can be provided. Then, an imaging method using this ⁇ -synuclein aggregate binding agent can be provided. Also, novel compounds that can be used as alpha-synuclein aggregate binding agents or other uses can be provided.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Neurosurgery (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Epidemiology (AREA)
  • Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Optics & Photonics (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Provided is a compound that has high binding selectivity to α-synuclein aggregates. The present invention relates to a compound represented by formula (I), (II) or (III), a pharmaceutically acceptable salt thereof, or a solvate of the compound or pharmaceutically acceptable salt.

Description

新規化合物、αシヌクレイン凝集体結合剤及びその利用Novel compound, α-synuclein aggregate binding agent and use thereof
 本発明は、新規化合物、αシヌクレイン凝集体結合剤及びその利用に関し、詳細には、新規化合物、当該新規化合物を含有するαシヌクレイン凝集体結合剤、αシヌクレイン凝集体の光学イメージング用組成物、αシヌクレイン凝集体の放射イメージング用組成物、脳内αシヌクレイン凝集体の光学イメージング方法、脳内αシヌクレイン凝集体の放射イメージング方法、及び、新規化合物を合成するための中間体に関する。 TECHNICAL FIELD The present invention relates to a novel compound, an α-synuclein aggregate binding agent and uses thereof, and in particular, a novel compound, an α-synuclein aggregate binding agent containing the novel compound, a composition for optical imaging of α-synuclein aggregate, α The present invention relates to a composition for radioimaging of brain α-synuclein aggregates, an optical imaging method of brain α-synuclein aggregates, a radioimaging method of brain α-synuclein aggregates, and intermediates for synthesizing novel compounds.
 αシヌクレインは、ニューロンのシナプス等に局在するタンパク質である。αシヌクレインの凝集体(以下、αシヌクレイン凝集体という)は、パーキンソン病、レビー小体型認知症(DLB)、多系統萎縮症(MSA)の中核病理を形成し、神経変性と密接な因果関係を有すると考えられている。こうした疾患の確定診断は、剖検脳の病理解析でαシヌクレイン凝集体(本明細書中「αシヌクレイン病変」ともいう)の存在を指標として行われるため、生前に診断を確定することはできない。しかしながら、生体脳でαシヌクレイン凝集体を可視化できれば、早期からこれらの疾患の確定診断に近い情報を得ることが可能になる。また、疾患モデル動物の生体脳でαシヌクレイン凝集体を可視化できれば、経時的なイメージング等により、αシヌクレイン凝集体を標的とする治療又は予防薬の候補物質の薬効評価にも役立ちうる。 α-Synuclein is a protein localized in synapses of neurons. Aggregates of α-synuclein (hereinafter referred to as α-synuclein aggregates) form the core pathology of Parkinson's disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA), and have a close causal relationship with neurodegeneration. are considered to have Definitive diagnosis of such diseases is made by pathological analysis of the brain at autopsy using the presence of α-synuclein aggregates (also referred to herein as "α-synuclein lesions") as an indicator, and thus the diagnosis cannot be confirmed before death. However, if α-synuclein aggregates can be visualized in the living brain, it will be possible to obtain information close to definitive diagnosis of these diseases at an early stage. In addition, if α-synuclein aggregates can be visualized in the living brains of disease model animals, it will be useful for efficacy evaluation of therapeutic or preventive drug candidate substances that target α-synuclein aggregates by imaging over time.
 従来生体脳でαシヌクレインに結合することが示されているPET(ポジトロン断層撮影法)プローブとして、[11C]BF-227がある(非特許文献1及び2)。しかしながら[11C]BF-227は、αシヌクレイン凝集体への結合親和性が十分ではなく、上記の疾患のうち、MSA患者の一部でαシヌクレイン病変が検出できるにとどまっている。また、[11C]BF-227は、非特異的な脳内集積という問題、及びアミロイドβ凝集体へ結合するためαシヌクレイン凝集体への結合選択性が低いという問題がある。 [ 11 C]BF-227 is a PET (positron emission tomography) probe that has been shown to bind to α-synuclein in the living brain (Non-Patent Documents 1 and 2). However, [ 11 C]BF-227 does not have sufficient binding affinity to α-synuclein aggregates, and among the above diseases, α-synuclein lesions can only be detected in some MSA patients. [ 11 C]BF-227 also has problems of non-specific accumulation in the brain and low binding selectivity to α-synuclein aggregates because it binds to amyloid β aggregates.
 ところで、発明者らは、脳内に蓄積したタウタンパク質をイメージング(画像化)するための化合物を開発した(特許文献1参照)。特許文献1に記載の化合物は、脳内に蓄積したタウタンパク質をイメージングできるため、特許文献1の技術は、タウタンパク質の蓄積により生じる疾患、例えばアルツハイマー病(AD)の治療及び予防等に役立つ。しかし、特許文献1にはαシヌクレイン凝集体への結合について記載されていない。 By the way, the inventors have developed a compound for imaging tau protein accumulated in the brain (see Patent Document 1). Since the compound described in Patent Document 1 can image tau protein accumulated in the brain, the technique of Patent Document 1 is useful for the treatment and prevention of diseases caused by the accumulation of tau protein, such as Alzheimer's disease (AD). However, Patent Document 1 does not describe binding to α-synuclein aggregates.
国際公開第2014/097474号WO2014/097474 国際公開第2020/174963号WO2020/174963
 本発明は、上記の状況に鑑みてなされたものであり、αシヌクレイン凝集体への結合選択性が高いαシヌクレイン凝集体結合剤、このαシヌクレイン凝集体結合剤を用いたイメージング方法、及びαシヌクレイン凝集体結合剤に用いることができる新規化合物を提供することを目的とする。 The present invention has been made in view of the above circumstances, and provides an α-synuclein aggregate-binding agent with high binding selectivity to α-synuclein aggregates, an imaging method using this α-synuclein aggregate-binding agent, and α-synuclein It is an object of the present invention to provide novel compounds that can be used as aggregate binders.
 本発明者らは、特定の構造を有する化合物がαシヌクレイン凝集体への結合選択性が高いことを見出し、さらに検討を重ねて、本発明を完成するに至った。より具体的には、本発明は以下のものを提供する。 The present inventors have found that a compound having a specific structure has high binding selectivity to α-synuclein aggregates, and after further investigation, have completed the present invention. More specifically, the present invention provides the following.
 本発明の一態様は、下記式(I)、(II)又は(III)で表される化合物、その医薬として許容し得る塩、又はその溶媒和物である。
Figure JPOXMLDOC01-appb-C000004
One aspect of the present invention is a compound represented by formula (I), (II) or (III) below, or a pharmaceutically acceptable salt or solvate thereof.
Figure JPOXMLDOC01-appb-C000004
 本発明によれば、αシヌクレイン凝集体への結合選択性が高いαシヌクレイン凝集体結合剤を提供することができる。 According to the present invention, an α-synuclein aggregate-binding agent with high binding selectivity to α-synuclein aggregates can be provided.
DLB患者脳及びAD患者脳の蛍光顕微鏡測定結果を示す図である。FIG. 4 shows fluorescence microscope measurement results of DLB patient brains and AD patient brains. 病変領域及び病変非形成領域の蛍光輝度の定量結果を示す図である。FIG. 10 is a diagram showing quantitative results of fluorescence intensity of a lesion area and a lesion-free area; αシヌクレイン線維接種マウスの蛍光顕微鏡測定結果を示す図である。FIG. 3 shows fluorescence microscope measurement results of α-synuclein fiber-inoculated mice. αシヌクレイン線維接種マウスの二光子レーザースキャニング蛍光顕微鏡検査結果を示す図である。FIG. 2 shows the results of two-photon laser scanning fluorescence microscopy of α-synuclein fiber-inoculated mice.
 以下、本発明の一実施形態について説明する。なお、本明細書において、「A及び/又はB」とは、A及びBの少なくとも一方を意図している。 An embodiment of the present invention will be described below. In addition, in this specification, "A and/or B" intends at least one of A and B.
 [定義]
 用語「医薬として許容し得る塩」とは、哺乳動物、特にヒトに対して有害でない塩を指す。医薬として許容し得る塩は、無機酸若しくは無機塩基、又は有機酸若しくは有機塩基を含む、無毒性の酸又は塩基を用いて形成することができる。医薬として許容し得る塩の例を挙げると、アルミニウム、カルシウム、リチウム、マグネシウム、カリウム、ナトリウム及び亜鉛などから形成される金属塩、又はリジン、N,N’-ジベンジルエチレンジアミン、クロロプロカイン、コリン、ジエタノールアミン、エチレンジアミン、メグルミン(N-メチルグルカミン)及びプロカインなどから形成される有機塩などがある。また、医薬として許容し得る塩は、酸付加塩及び塩基付加塩を包含する。
[definition]
The term "pharmaceutically acceptable salt" refers to salts that are not harmful to mammals, especially humans. Pharmaceutically acceptable salts can be formed with non-toxic acids or bases, including inorganic acids or bases, or organic acids or bases. Examples of pharmaceutically acceptable salts include metal salts formed from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc, or lysine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, and organic salts formed from diethanolamine, ethylenediamine, meglumine (N-methylglucamine), procaine, and the like. Pharmaceutically acceptable salts also include acid addition salts and base addition salts.
 用語「医薬として許容し得る担体」とは、生理食塩水溶液、液体若しくは固体の充填剤、希釈剤、溶媒、又は封入材料などの医薬として許容し得る材料、組成物、又はビヒクルを意味する。医薬として許容し得る担体の例を挙げると、水、食塩水、生理食塩水又はリン酸緩衝食塩水(PBS)、塩化ナトリウム注射液、リンゲル注射液、等張性デキストロース注射液、無菌水注射液、デキストロース、及び乳酸リンゲル注射液などがある。 The term "pharmaceutically acceptable carrier" means a pharmaceutically acceptable material, composition, or vehicle such as a saline solution, liquid or solid filler, diluent, solvent, or encapsulating material. Examples of pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. , dextrose, and Lactated Ringer's Injection.
 用語「有効量」とは、目的の効果を得ることができる、化合物又は組成物の量をいう。例えば、一部の実施態様において、有効量は、αシヌクレイン凝集体等の脳内に蓄積する物質の光学イメージング又は放射イメージングが可能な、化合物又は組成物の量をいう。 The term "effective amount" refers to the amount of a compound or composition that can achieve the desired effect. For example, in some embodiments, an effective amount refers to that amount of a compound or composition that allows for optical or radioimaging of substances that accumulate in the brain, such as alpha-synuclein aggregates.
 用語「溶媒和物」とは、化合物に対する1つ又は複数の溶媒分子の会合により形成される含溶媒化合物を意味する。溶媒和物は、例えば、一溶媒和物、二溶媒和物、三溶媒和物、及び四溶媒和物を含む。また、溶媒和物は、水和物を含む。 The term "solvate" means a solvate formed by the association of one or more solvent molecules with the compound. Solvates include, for example, mono-, di-, tri-, and tetra-solvates. Solvates also include hydrates.
 用語「水和物」とは、非共有結合性分子間力によって拘束された化学量論的又は非化学量論的量の水をさらに含む化合物又はその塩を意味する。水和物は、例えば、一水和物、二水和物、三水和物、及び四水和物などを含む。 The term "hydrate" means a compound or salt thereof further containing a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces. Hydrates include, for example, monohydrates, dihydrates, trihydrates, tetrahydrates, and the like.
 用語「治療」とは、疾患又は状態の進行、重症度及び/又は持続期間を低減すること又は寛解することを意味する。 The term "treatment" means reducing or ameliorating the progression, severity and/or duration of a disease or condition.
 用語「予防」とは、所定の疾患又は状態を獲得する又は進行させる危険の低減、或いは、所定の疾患又は条件の1つ又は複数の症状の再発、開始、又は進行の低減又は抑制を意味する。 The term "prevention" means reducing the risk of acquiring or developing a given disease or condition, or reducing or inhibiting the recurrence, onset, or progression of one or more symptoms of a given disease or condition. .
 用語「結合性」とは、ある特定のタンパク質凝集体に対する化合物の結合強度を意味する。 The term "binding" means the binding strength of a compound to a specific protein aggregate.
 用語「結合選択性」とは、化合物の、ある特定のタンパク質凝集体に対する結合性と、その他のタンパク質凝集体に対する結合性を比較したとき、それらの結合性に差がある(ある特定のタンパク質凝集体に対する結合性がその他のタンパク質凝集体に対する結合性より高い又は低い)ことを指す。「結合選択性が高い」とは、上記の結合性の差が大きいことを意味する。例えば、ある化合物が「αシヌクレイン凝集体への結合選択性が高い」とは、当該化合物のαシヌクレイン凝集体に対する結合性とその他のタンパク質凝集体に対する結合性には大きな差があり、αシヌクレイン凝集体により高い結合性を有していることを示す。 The term "binding selectivity" means that there is a difference in the binding properties of a compound when comparing the binding properties of a compound to a specific protein aggregate and the binding properties of other protein aggregates (specific protein aggregates). binding to aggregates is higher or lower than binding to other protein aggregates). “High binding selectivity” means that the difference in the above binding properties is large. For example, when a compound "has high selectivity for binding to α-synuclein aggregates", it means that there is a large difference between the binding ability of the compound to α-synuclein aggregates and the binding ability to other protein aggregates. It shows that the aggregates have higher binding properties.
 [化合物]
 一実施形態において、本発明は、下記構造式(I)で表される(E)-1-フルオロ-3-((2-(4-(5-メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オール、下記構造式(II)で表される(E)-1-フルオロ-3-((2-(4-(6-メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オール、又は下記構造式(III)で表される(E)-1-フルオロ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロパン-2-オール、その医薬として許容し得る塩、又はその溶媒和物を提供する。
Figure JPOXMLDOC01-appb-C000005
 本明細書において、式(I)、式(II)及び式(III)で表される化合物をそれぞれ「化合物(I)」、「化合物(II)」及び「化合物(III)」ともいう。
[Compound]
In one embodiment, the present invention provides (E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazin-2-yl)buta- 1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol, (E)-1-fluoro-3- represented by the following structural formula (II) ((2-(4-(6-methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol , or (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-ene-3-) represented by the following structural formula (III) In-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propan-2-ol, a pharmaceutically acceptable salt thereof, or a solvate thereof.
Figure JPOXMLDOC01-appb-C000005
In this specification, the compounds represented by formula (I), formula (II) and formula (III) are also referred to as "compound (I)", "compound (II)" and "compound (III)" respectively.
 一実施態様において、化合物(I)、化合物(II)及び化合物(III)は、1個又はそれ以上の原子が該原子の放射性同位体である化合物、その塩、又はその溶媒和物である。放射性同位体は、15O、13N、11C、及び18Fなどからなる群から選択されるが、特に限定されない。好ましくは、放射性同位体は、11C又は18Fである。このうち、11Cの半減期が約20分であり、18Fの半減期が約110分であることを考慮すれば、18Fで標識した化合物の方が、商業的利用価値が高いものと考えられる。したがって、最も好ましくは、放射性同位体は、18Fである。 In one embodiment, compound (I), compound (II) and compound (III) are compounds wherein one or more atoms are radioactive isotopes of said atoms, salts thereof, or solvates thereof. Radioisotopes are selected from the group consisting of 15 O, 13 N, 11 C, and 18 F, but are not particularly limited. Preferably, the radioisotope is 11 C or 18 F. Considering that 11 C has a half-life of about 20 minutes and that of 18 F has a half-life of about 110 minutes, the 18 F-labeled compound is considered to have a higher commercial utility value. Conceivable. Therefore, most preferably the radioisotope is 18F .
 好ましくは、ピラジン環若しくはピリジン環に結合しているメチルアミノ基、及びベンゾチアゾール環に結合している3-フルオロ-2-ヒドロキシプロピルアミノ基(-NH-CH-CH(OH)-CHF)若しくはチアゾロピリジン環に結合している3-フルオロ-2-ヒドロキシプロポキシ基(-O-CH-CH(OH)-CHF)の少なくとも一方が、放射性同位体を含む基である。より好ましくは、3-フルオロ-2-ヒドロキシプロピルアミノ基又は3-フルオロ-2-ヒドロキシプロポキシ基が、放射性同位体を含む基である。さらに好ましくは、3-フルオロ-2-ヒドロキシプロピルアミノ基又は3-フルオロ-2-ヒドロキシプロポキシ基におけるフッ素原子が放射性同位体である。 Preferably, a methylamino group bonded to a pyrazine ring or a pyridine ring, and a 3-fluoro-2-hydroxypropylamino group (-NH-CH 2 -CH(OH)-CH 2 bonded to a benzothiazole ring) At least one of F) or a 3-fluoro-2-hydroxypropoxy group (—O—CH 2 —CH(OH)—CH 2 F) bonded to the thiazolopyridine ring is a group containing a radioactive isotope. . More preferably, a 3-fluoro-2-hydroxypropylamino group or a 3-fluoro-2-hydroxypropoxy group is a group containing a radioisotope. More preferably, the fluorine atom in the 3-fluoro-2-hydroxypropylamino group or 3-fluoro-2-hydroxypropoxy group is a radioactive isotope.
 一実施態様において、放射性同位体を含む化合物(I)は、好ましくは、[18F]-(E)-1-フルオロ-3-((2-(4-(5-メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オールである。 In one embodiment, the compound (I) containing a radioisotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazine-2 -yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol.
 一実施態様において、放射性同位体を含む化合物(II)は、好ましくは、[18F]-(E)-1-フルオロ-3-((2-(4-(6-メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オールである。 In one embodiment, the compound (II) containing a radioisotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(6-methylamino)pyridine-3 -yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-yl)amino)propan-2-ol.
 一実施態様において、放射性同位体を含む化合物(III)は、好ましくは、[18F]-(E)-1-フルオロ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロパン-2-オールである。 In one embodiment, the compound (III) containing a radioactive isotope is preferably [ 18 F]-(E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazine- 2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propan-2-ol.
 [中間体]
 下記の化合物の製造方法に示されている通り、下記式(IV)で表される化合物は、化合物(I)及び化合物(II)の製造中間体化合物である。また、下記式(V)で表される化合物は、化合物(III)の製造中間体化合物である。また、下記式(VI)で表される化合物は、(I)及び化合物(II)の製造中間体化合物である。本明細書において、これら製造中間体化合物を、「中間体化合物」又は単に「中間体」ともいう。
Figure JPOXMLDOC01-appb-C000006
 式(IV)及び式(V)中、Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基(トシル基)又はメタンスルホニル基である。Rは、テトラヒドロ-2H-ピラン-2-イル基又はメトキシメチル基である。Rは、水素原子、tert-ブトキシカルボニル(Boc)基又は2,4-ジメトキシベンジル基である。
[Intermediate]
As shown in the compound production method below, the compound represented by the following formula (IV) is an intermediate compound for the production of compound (I) and compound (II). Moreover, the compound represented by the following formula (V) is a production intermediate compound of compound (III). In addition, the compound represented by the following formula (VI) is an intermediate compound for the production of (I) and compound (II). In this specification, these production intermediate compounds are also referred to as "intermediate compounds" or simply "intermediates".
Figure JPOXMLDOC01-appb-C000006
In formulas (IV) and (V), R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group (tosyl group) or a methanesulfonyl group. R 2 is a tetrahydro-2H-pyran-2-yl group or a methoxymethyl group. R 4 is a hydrogen atom, a tert-butoxycarbonyl (Boc) group or a 2,4-dimethoxybenzyl group.
 式(IV)中、Rは、水素原子、tert-ブトキシカルボニル基又は2,4-ジメトキシベンジル基である。Xは窒素原子又は置換されていない炭素原子である。本明細書において「置換されていない炭素原子」は、CHを示す。
Figure JPOXMLDOC01-appb-C000007
 式(VI)中、
 Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基又はメタンスルホニル基であり、
 R4’は、水素原子又はtert-ブトキシカルボニル基であり、
 Xは窒素原子(N)又は置換されていない炭素原子(CH)である。
In formula (IV), R 3 is a hydrogen atom, tert-butoxycarbonyl group or 2,4-dimethoxybenzyl group. X is a nitrogen atom or an unsubstituted carbon atom. As used herein, "unsubstituted carbon atom" refers to CH.
Figure JPOXMLDOC01-appb-C000007
In formula (VI),
R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group,
R 4' is a hydrogen atom or a tert-butoxycarbonyl group,
X is a nitrogen atom (N) or an unsubstituted carbon atom (CH).
 これら中間体化合物は、化合物(I)及び化合物(II)の合成又は化合物(III)の合成、さらに放射性同位体標識された化合物(I)及び化合物(II)の合成又は放射性同位体標識された化合物(III)の合成に好適に用いられる。また、これら中間体化合物は、塩であってもよい。 These intermediate compounds can be used to synthesize compound (I) and compound (II) or synthesize compound (III), further synthesize radioisotope-labeled compound (I) and compound (II), or radioisotope-labeled It is preferably used for synthesizing compound (III). Also, these intermediate compounds may be salts.
 化合物(I)、化合物(II)又は化合物(III)が、各々立体異性体(光学異性体及び回転異性体を含む)、互変異性体、又は極性体等の各種異性体を含有する場合には、これらの異性体も、化合物(I)、化合物(II)又は化合物(III)に包含される。これらの異性体は、公知の合成手法、分離手法により、それぞれを単品として得ることができる。化合物(IV)、化合物(V)及び化合物(VI)もまた、各種異性体を包含し得る。 When compound (I), compound (II) or compound (III) contains various isomers such as stereoisomers (including optical isomers and rotational isomers), tautomers, or polar isomers, respectively are also included in compound (I), compound (II) or compound (III). These isomers can be obtained as single products by known synthetic techniques and separation techniques. Compound (IV), compound (V) and compound (VI) may also include various isomers.
 化合物(I)~化合物(VI)はまた、公知の結晶化法により製造される結晶であってもよい。 Compounds (I) to (VI) may also be crystals produced by known crystallization methods.
 〔化合物の製造方法〕
 化合物(I)~化合物(VI)は、以下に示す製造法に準ずる方法に従い製造することができる。また、所望により、脱保護反応、アミド化反応、ウレア化反応、アルキル化反応、光延反応、酸化反応、還元反応、ハロゲン化反応、カップリング反応、カルボアニオンによる求核付加反応、Grignard反応、脱水反応などを各々、単独又はその二つ以上を組み合わせて行うことにより化合物(I)~化合物(VI)を製造することができる。また、各反応における溶媒、試薬及び温度等の反応条件は、当業者の技術常識に基づき適宜設定すればよい。官能基の保護及び脱保護反応は、公知の反応方法、参考例又は実施例に記載された方法に準じて行われ、保護基としては慣用のものが用いられる。
[Method for producing compound]
Compounds (I) to (VI) can be produced according to the production method shown below. Further, if desired, deprotection reaction, amidation reaction, urea reaction, alkylation reaction, Mitsunobu reaction, oxidation reaction, reduction reaction, halogenation reaction, coupling reaction, nucleophilic addition reaction with carbanion, Grignard reaction, dehydration. Compounds (I) to (VI) can be produced by performing each reaction alone or in combination of two or more thereof. In addition, reaction conditions such as solvents, reagents and temperatures in each reaction may be appropriately set based on the common technical knowledge of those skilled in the art. Protection and deprotection reactions of functional groups are carried out according to known reaction methods and methods described in Reference Examples or Examples, and conventional protecting groups are used.
 下記の製造法中、特に指定しない限り、用いられる各記号の意味は先述と同様である。 Unless otherwise specified, the meaning of each symbol used in the manufacturing method below is the same as described above.
 また、下記の製造法の説明では、各製造法で示される各スキーム中、(1)、(2)等で示す化合物を、化合物(1)、化合物(2)等と称する。
 [1.化合物(I)及び化合物(II)並びにその中間体である化合物(IV)の製造方法]
 (製造法A)
 一態様として、化合物(I)及び化合物(II)は、製造スキーム1で示す以下の方法で製造することができる。
In addition, in the description of the production methods below, the compounds indicated by (1), (2), etc. in each scheme shown in each production method are referred to as compound (1), compound (2), etc.
[1. Method for producing compound (I), compound (II), and its intermediate compound (IV)]
(Manufacturing method A)
In one aspect, compound (I) and compound (II) can be produced by the following method shown in Production Scheme 1.
 (製造スキーム1)
Figure JPOXMLDOC01-appb-C000008
 製造スキーム1中、TBSはtert-ブチルジメチルシリル基を示す。
(Manufacturing scheme 1)
Figure JPOXMLDOC01-appb-C000008
In Production Scheme 1, TBS represents a tert-butyldimethylsilyl group.
 化合物(3)は、化合物(2)のオキシラン開環反応による、化合物(1)のアルキル化反応により製造することができる。 Compound (3) can be produced by an alkylation reaction of compound (1) through an oxirane ring-opening reaction of compound (2).
 化合物(4)は、塩化tert-ブチルジメチルシリルによる化合物(3)の水酸基保護反応により製造することができる。 Compound (4) can be produced by hydroxyl-protecting compound (3) with tert-butyldimethylsilyl chloride.
 化合物(5)は、二炭酸ジ-tert-ブチルによる化合物(4)のアミノ基保護反応により製造することができる。 Compound (5) can be produced by amino group-protecting reaction of compound (4) with di-tert-butyl dicarbonate.
 化合物(6)は、N,N-ジメチルホルムアミドなどによる化合物(5)のホルミル化反応により製造することができる。 Compound (6) can be produced by formylating compound (5) with N,N-dimethylformamide or the like.
 化合物(7)は、(ブロモメチル)トリフェニルホスホニウムブロミドを用いた化合物(6)のビニルブロモ化反応により製造することができる。 Compound (7) can be produced by a vinyl bromination reaction of compound (6) using (bromomethyl)triphenylphosphonium bromide.
 化合物(9)は、化合物(7)と後述する化合物(8)との薗頭反応などにより製造することができる。 Compound (9) can be produced by the Sonogashira reaction between compound (7) and compound (8) described below.
 化合物(I)及び化合物(II)は、化合物(9)の脱保護反応により製造することができる。 Compound (I) and compound (II) can be produced by a deprotection reaction of compound (9).
 製造法Aで用いられる化合物(8)は、製造スキーム2で示す以下の方法で化合物(10)から製造することができる。 Compound (8) used in Production Method A can be produced from Compound (10) by the following method shown in Production Scheme 2.
 (製造スキーム2)
Figure JPOXMLDOC01-appb-C000009
 製造スキーム2中、Halはハロゲン原子(例えば、臭素原子又はヨウ素原子)を示し、TMSはトリメチルシリル基を示す。
(Manufacturing scheme 2)
Figure JPOXMLDOC01-appb-C000009
In Production Scheme 2, Hal represents a halogen atom (eg, bromine atom or iodine atom), and TMS represents a trimethylsilyl group.
 化合物(11)は、二炭酸ジ-tert-ブチルによる化合物(10)のアミノ基保護(ジBoc化)反応により製造することができる。 Compound (11) can be produced by amino group-protecting (di-Boc) reaction of compound (10) with di-tert-butyl dicarbonate.
 化合物(12)は、メタノールなどの溶媒中、炭酸カリウムなどの塩基による化合物(11)における1つのBoc基の脱保護(モノBoc化)反応により製造することができる。 Compound (12) can be produced by a deprotection (mono-Boc) reaction of one Boc group in compound (11) with a base such as potassium carbonate in a solvent such as methanol.
 化合物(13)は、ヨードメタンなどによる化合物(12)のメチル化反応により製造することができる。 Compound (13) can be produced by a methylation reaction of compound (12) with iodomethane or the like.
 化合物(15)は、化合物(13)とエチニルトリメチルシラン(14)との薗頭反応により製造することができる。 Compound (15) can be produced by a Sonogashira reaction between compound (13) and ethynyltrimethylsilane (14).
 化合物(8)は、化合物(15)のTMS基の脱保護反応により製造することができる。 Compound (8) can be produced by a deprotection reaction of the TMS group of compound (15).
 (製造法B)
 別の態様として、化合物(I)及び化合物(II)は、製造スキーム3で示す以下の方法で製造することもできる。
(Manufacturing method B)
In another aspect, compound (I) and compound (II) can also be produced by the following method shown in Production Scheme 3.
 (製造スキーム3)
Figure JPOXMLDOC01-appb-C000010
 化合物(17)は、N,N-ジメチルホルムアミドなどによる化合物(16)のホルミル化反応により製造することができる。
(Manufacturing scheme 3)
Figure JPOXMLDOC01-appb-C000010
Compound (17) can be produced by formylating compound (16) with N,N-dimethylformamide or the like.
 化合物(18)は、(ブロモメチル)トリフェニルホスホニウムブロミドを用いた化合物(17)のビニルブロモ化反応により製造することができる。 Compound (18) can be produced by a vinyl bromination reaction of compound (17) using (bromomethyl)triphenylphosphonium bromide.
 化合物(19)は、化合物(18)と化合物(8)との薗頭反応などにより製造することができる。 Compound (19) can be produced by the Sonogashira reaction of compound (18) and compound (8).
 化合物(20)は、化合物(19)の脱保護反応により製造することができる。 Compound (20) can be produced by a deprotection reaction of compound (19).
 化合物(I)及び化合物(II)は、化合物(2)のオキシラン開環反応による、化合物(20)のアルキル化反応により製造することができる。 Compound (I) and compound (II) can be produced by an oxirane ring-opening reaction of compound (2) and an alkylation reaction of compound (20).
 また、化合物(2)のオキシラン開環反応による、化合物(20)のアルキル化反応において、フッ素原子が18Fである化合物(2)、すなわち[18F]2-(フルオロメチル)オキシラン(CAS[123436-06-6])を用いることにより、3-フルオロ-2-ヒドロキシプロピルアミノ基におけるフッ素原子が放射性同位体である化合物(I)及び化合物(II)を製造することができる。ここで、[18F]2-(フルオロメチル)オキシランの合成は、[18F]フッ化物イオンを用いたグリシジルトシレートの求核置換及び蒸留による精製により実施することができる。[18F]2-(フルオロメチル)オキシランの合成の一例を下記に示す。
Figure JPOXMLDOC01-appb-C000011
Further, in the alkylation reaction of compound (20) by the oxirane ring-opening reaction of compound (2), compound (2) having a fluorine atom of 18 F, that is, [ 18 F]2-(fluoromethyl)oxirane (CAS[ 123436-06-6]), compounds (I) and compounds (II) in which the fluorine atom in the 3-fluoro-2-hydroxypropylamino group is a radioactive isotope can be produced. Here, [ 18 F]2-(fluoromethyl)oxirane can be synthesized by nucleophilic substitution of glycidyl tosylate with [ 18 F]fluoride ion and purification by distillation. An example of the synthesis of [ 18 F]2-(fluoromethyl)oxirane is shown below.
Figure JPOXMLDOC01-appb-C000011
 (製造法C)
 さらに別の態様として、R及びRがBoc基である化合物(IV)(化合物(IV-i))は、製造スキーム4で示す以下の方法で製造することができる。
(Manufacturing method C)
In still another embodiment, compound (IV) (compound (IV-i)) in which R 3 and R 4 are Boc groups can be produced by the following method shown in Production Scheme 4.
 (製造スキーム4)
Figure JPOXMLDOC01-appb-C000012
 製造スキーム4中、Rはハロゲン原子(例えば、臭素原子又はヨウ素原子)又は4-ニトロベンゼンスルホニルオキシ基などの脱離基を示す。
(Manufacturing scheme 4)
Figure JPOXMLDOC01-appb-C000012
In Preparation Scheme 4, R 5 represents a halogen atom (eg, bromine atom or iodine atom) or a leaving group such as a 4-nitrobenzenesulfonyloxy group.
 化合物(IV-i)は、後述する化合物(21)による化合物(19)のアルキル化反応などにより製造することができる。 Compound (IV-i) can be produced by an alkylation reaction of compound (19) with compound (21), which will be described later, or the like.
 製造法Cで用いられる化合物(21)においてRがハロゲン原子であり、Rがトシル基または4-ニトロベンゼンスルホニル基である化合物(21a)は、製造スキーム5で示す以下の方法で化合物(22)から製造することができる。 Compound (21a) in which R 5 is a halogen atom and R 1 is a tosyl group or a 4-nitrobenzenesulfonyl group in Compound (21) used in Production Method C can be prepared by the following method shown in Production Scheme 5 to Compound (22 ).
 (製造スキーム5)
Figure JPOXMLDOC01-appb-C000013
 製造スキーム5中、R1aはトシル基または4-ニトロベンゼンスルホニル基を示し、Halはハロゲン原子(例えば、ヨウ素原子)を示す。
(Manufacturing scheme 5)
Figure JPOXMLDOC01-appb-C000013
In Production Scheme 5, R 1a represents a tosyl group or a 4-nitrobenzenesulfonyl group, and Hal represents a halogen atom (eg, iodine atom).
 化合物(23)は、化合物(22)のスルホニル化反応により製造することができる。 Compound (23) can be produced by a sulfonylation reaction of compound (22).
 化合物(21a)は、テトラヒドロピラニル(THP)基、メトキシメチル(MOM)基などによる化合物(23)の水酸基保護反応により製造することができる。 Compound (21a) can be produced by a hydroxyl group-protecting reaction of compound (23) with a tetrahydropyranyl (THP) group, a methoxymethyl (MOM) group, or the like.
 (製造法C’)
 化合物(I)及び(II)は、製造中間体化合物(IV)から以下の製造スキーム4’で示す方法で製造することができる。
(Manufacturing method C')
Compounds (I) and (II) can be produced from production intermediate compound (IV) by the method shown in Production Scheme 4' below.
 (製造スキーム4’)
Figure JPOXMLDOC01-appb-C000014
 化合物(I)及び(II)は、製造中間体化合物(IV)のフッ化カリウムなどによるフッ素化反応後の脱保護反応により製造することができる。また、[18F]フッ化カリウムなどを用いることにより、放射性同位体である化合物(I)及び化合物(II)を製造することができる。
 [2.化合物(III)及びその中間体である化合物(V)の製造方法]
 (製造法D)
 一態様として、化合物(III)は、製造スキーム6で示す以下の方法で製造することができる。
(Manufacturing scheme 4')
Figure JPOXMLDOC01-appb-C000014
Compounds (I) and (II) can be produced by a deprotection reaction after a fluorination reaction of production intermediate compound (IV) with potassium fluoride or the like. Further, by using [ 18 F]potassium fluoride or the like, compound (I) and compound (II), which are radioactive isotopes, can be produced.
[2. Method for producing compound (III) and its intermediate compound (V)]
(Manufacturing method D)
In one aspect, compound (III) can be produced by the following method shown in Production Scheme 6.
 (製造スキーム6)
Figure JPOXMLDOC01-appb-C000015
 化合物(32)は、化合物(31)を用いた環化反応により製造することができる。
(Manufacturing scheme 6)
Figure JPOXMLDOC01-appb-C000015
Compound (32) can be produced by a cyclization reaction using compound (31).
 化合物(33)は、化合物(32)のブロモ化反応により製造することができる。 Compound (33) can be produced by a bromination reaction of compound (32).
 化合物(34)は、化合物(33)と亜リン酸トリエチルとの反応により製造することができる。 Compound (34) can be produced by reacting compound (33) with triethyl phosphite.
 化合物(35)は、3,4-ジメトキシベンジルアルコールによる化合物(34)のアルコキシル化反応により製造することができる。 Compound (35) can be produced by an alkoxylation reaction of compound (34) with 3,4-dimethoxybenzyl alcohol.
 化合物(37)は、化合物(35)と後述する化合物(36)とのホーナー・ワズワース・エモンズ反応(HWE反応)により製造することができる。 Compound (37) can be produced by Horner-Wadsworth-Emmons reaction (HWE reaction) between compound (35) and compound (36) described below.
 化合物(38)は、化合物(37)の脱保護反応により製造することができる。 Compound (38) can be produced by a deprotection reaction of compound (37).
 化合物(III)は、化合物(39)のオキシラン開環反応による、化合物(38)へのアルキル化反応により製造することができる。 Compound (III) can be produced by an oxirane ring-opening reaction of compound (39) and an alkylation reaction to compound (38).
 製造法Dで用いられる化合物(36)は、製造スキーム7で示す以下の方法で化合物(40)から製造することができる。 Compound (36) used in Production Method D can be produced from Compound (40) by the following method shown in Production Scheme 7.
 また、化合物(39)のオキシラン開環反応による、化合物(38)のアルキル化反応において、フッ素原子が18Fである化合物(39)、すなわち[18F]2-(フルオロメチル)オキシランを用いることにより、3-フルオロ-2-ヒドロキシプロポキシ基におけるフッ素原子が放射性同位体である化合物(III)を製造することができる。 In addition, in the alkylation reaction of compound (38) by the oxirane ring-opening reaction of compound (39), compound (39) in which the fluorine atom is 18 F, that is, [ 18 F]2-(fluoromethyl)oxirane is used. can prepare compound (III) in which the fluorine atom in the 3-fluoro-2-hydroxypropoxy group is a radioactive isotope.
 (製造スキーム7)
Figure JPOXMLDOC01-appb-C000016
 化合物(41)は、二炭酸ジ-tert-ブチルによる化合物(40)のアミノ基保護(ジBoc化)反応により製造することができる。
(Manufacturing scheme 7)
Figure JPOXMLDOC01-appb-C000016
Compound (41) can be produced by amino group-protecting (di-Boc) reaction of compound (40) with di-tert-butyl dicarbonate.
 化合物(42)は、メタノールなどの溶媒中、炭酸カリウムなどの塩基による化合物(41)における1つのBoc基の脱保護(モノBoc化)反応により製造することができる。 Compound (42) can be produced by a deprotection (mono-Boc) reaction of one Boc group in compound (41) with a base such as potassium carbonate in a solvent such as methanol.
 化合物(43)は、ヨードメタンなどによる化合物(42)のメチル化反応により製造することができる。 Compound (43) can be produced by a methylation reaction of compound (42) with iodomethane or the like.
 化合物(45)は、化合物(43)とプロパルギルアルコール(44)との薗頭反応により製造することができる。 Compound (45) can be produced by a Sonogashira reaction between compound (43) and propargyl alcohol (44).
 化合物(36)は、化合物(45)の酸化反応により製造することができる。 Compound (36) can be produced by oxidation reaction of compound (45).
 (製造法E)
 別の態様として、RがHである化合物(V)(化合物(V-i))は、製造スキーム8で示す以下の方法で製造することができる。
(Manufacturing method E)
In another embodiment, compound (V) (compound (Vi)) in which R 4 is H can be produced by the following method shown in Production Scheme 8.
 (製造スキーム8)
Figure JPOXMLDOC01-appb-C000017
 製造スキーム8中、TBSはtert-ブチルジメチルシリル基を示す。
(Manufacturing scheme 8)
Figure JPOXMLDOC01-appb-C000017
In Production Scheme 8, TBS represents a tert-butyldimethylsilyl group.
 化合物(47)は、化合物(38)と後述する化合物(46)との光延反応などのアルキル化反応により製造することができる。 Compound (47) can be produced by an alkylation reaction such as Mitsunobu reaction between compound (38) and compound (46) described below.
 化合物(48)は、化合物(47)の脱保護反応により製造することができる。 Compound (48) can be produced by a deprotection reaction of compound (47).
 化合物(V-i)は、化合物(48)の保護反応により製造することができる。 Compound (Vi) can be produced by protecting compound (48).
 製造法Eで用いられる化合物(46)において、Rがパラトルエンスルホニル基又は4-ニトロベンゼンスルホニル基である化合物(46a)は、製造スキーム9で示す以下の方法で化合物(49)から製造することができる。 In compound (46) used in production method E, compound (46a) in which R 1 is a p-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group can be produced from compound (49) by the following method shown in production scheme 9. can be done.
 (製造スキーム9)
Figure JPOXMLDOC01-appb-C000018
 製造スキーム9中、R1bはパラトルエンスルホニル基または4-ニトロベンゼンスルホニル基を示す。
(Manufacturing scheme 9)
Figure JPOXMLDOC01-appb-C000018
In Production Scheme 9, R 1b represents a p-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group.
 化合物(50)は、化合物(49)の水酸基保護反応により製造することができる。 Compound (50) can be produced by hydroxyl group protection reaction of compound (49).
 化合物(46a)は、化合物(50)の脱ベンジル反応により製造することができる。 Compound (46a) can be produced by a debenzylation reaction of compound (50).
 (製造法F)
 さらに別の態様として、化合物(V-i)は、製造スキーム10で示す以下の方法で製造することができる。
(Manufacturing method F)
In still another embodiment, compound (Vi) can be produced by the following method shown in Production Scheme 10.
 (製造スキーム10)
Figure JPOXMLDOC01-appb-C000019
 製造スキーム10中、Rはハロゲン原子(例えば、臭素原子又はヨウ素原子)、パラトルエンスルホニルオキシ基又は4-ニトロベンゼンスルホニルオキシ基などの脱離基を示す。
(Manufacturing scheme 10)
Figure JPOXMLDOC01-appb-C000019
In Preparation Scheme 10, R 5 represents a leaving group such as a halogen atom (eg, bromine atom or iodine atom), paratoluenesulfonyloxy group or 4-nitrobenzenesulfonyloxy group.
 化合物(V-i)は、後述する化合物(51)による、化合物(38)のアルキル化反応などにより製造することができる。 Compound (Vi) can be produced by, for example, alkylating compound (38) with compound (51) described below.
 製造法Fで用いられる化合物(51)においてRがハロゲン原子であり、Rがパラトルエンスルホニル基又は4-ニトロベンゼンスルホニル基である化合物は、上述の[2.化合物(I)及び化合物(II)並びにその中間体である化合物(IV)の製造方法]の製造スキーム5中の化合物(21a)であり、当該製造スキーム5に示す方法で製造することができる。 The compound (51) used in production method F in which R 5 is a halogen atom and R 1 is a para-toluenesulfonyl group or a 4-nitrobenzenesulfonyl group is the compound described in [2. Compound (21a) in Production Scheme 5 of Compound (I), Compound (II), and Compound (IV), which is an intermediate thereof], and can be produced by the method shown in Production Scheme 5.
 (製造法F’)
 化合物(III)は、製造中間体化合物(V)から以下の製造スキーム10’で示す方法で製造することができる。
(Manufacturing method F')
Compound (III) can be produced from production intermediate compound (V) by the method shown in the following production scheme 10'.
 (製造スキーム10’)
Figure JPOXMLDOC01-appb-C000020
 化合物(III)は、製造中間体化合物(V)のフッ化カリウムなどによるフッ素化反応後の脱保護反応により製造することができる。また、[18F]フッ化カリウムなどを用いることにより、放射性同位体である化合物(III)を製造することができる。
(Manufacturing scheme 10')
Figure JPOXMLDOC01-appb-C000020
Compound (III) can be produced by a deprotection reaction after a fluorination reaction of production intermediate compound (V) with potassium fluoride or the like. Further, by using [ 18 F]potassium fluoride or the like, compound (III), which is a radioactive isotope, can be produced.
 [3.化合物(I)及び化合物(II)の中間体である化合物(VI)の製造方法]
(製造法G)
 一態様として、Rがパラトルエンスルホニル基(トシル基)であり、R4’が水素原子である化合物(VI)(化合物(VI-i))は、製造スキーム11で示す以下の方法で製造することができる。
[3. Method for producing compound (VI), which is an intermediate of compound (I) and compound (II)]
(Manufacturing method G)
In one embodiment, the compound (VI) (compound (VI-i)) in which R 1 is a p-toluenesulfonyl group (tosyl group) and R 4' is a hydrogen atom is produced by the following method shown in Production Scheme 11. can do.
 (製造スキーム11)
Figure JPOXMLDOC01-appb-C000021
 化合物(53)は、化合物(52)のトシル化反応により製造することができる。
(Manufacturing scheme 11)
Figure JPOXMLDOC01-appb-C000021
Compound (53) can be produced by subjecting compound (52) to a tosylation reaction.
 化合物(54)は、化合物(53)の水酸基の酸化反応により製造することができる。 Compound (54) can be produced by oxidation reaction of the hydroxyl group of compound (53).
 化合物(55)は、化合物(20)と化合物(54)とのイミン形成反応により製造することができる。 Compound (55) can be produced by an imine formation reaction between compound (20) and compound (54).
 化合物(56)は、化合物(55)のイミノ基の還元反応により製造することができる。 Compound (56) can be produced by a reduction reaction of the imino group of compound (55).
 化合物(57)は、化合物(56)の脱保護反応により製造することができる。 Compound (57) can be produced by a deprotection reaction of compound (56).
 化合物(58)は、化合物(57)のカルボニル基の還元反応により製造することができる。 Compound (58) can be produced by a reduction reaction of the carbonyl group of compound (57).
 化合物(VI-i)は、化合物(58)のアミノ基と水酸基でのヘミアミナールエーテル化反応により製造することができる。 Compound (VI-i) can be produced by a hemiaminal etherification reaction between the amino group and hydroxyl group of compound (58).
 (製造法G’)
 化合物(I)及び(II)は、製造中間体化合物(VI)から以下の製造スキーム11’で示す方法で製造することができる。
(Manufacturing method G')
Compounds (I) and (II) can be produced from production intermediate compound (VI) by the method shown in the following production scheme 11'.
 (製造スキーム11’)
Figure JPOXMLDOC01-appb-C000022
 化合物(I)及び(II)は、製造中間体化合物(VI)のフッ化カリウムなどによるフッ素化反応後の脱保護反応により製造することができる。また、[18F]フッ化カリウムなどを用いることにより、放射性同位体である化合物(I)及び化合物(II)を製造することができる。
(Manufacturing scheme 11')
Figure JPOXMLDOC01-appb-C000022
Compounds (I) and (II) can be produced by a deprotection reaction after a fluorination reaction of production intermediate compound (VI) with potassium fluoride or the like. Further, by using [ 18 F]potassium fluoride or the like, compound (I) and compound (II), which are radioactive isotopes, can be produced.
 [αシヌクレイン凝集体結合剤]
 一実施形態において、本発明は、αシヌクレイン凝集体結合剤を提供する。本実施形態のαシヌクレイン凝集体結合剤(以下、結合剤とも記載する)は、化合物(I)、化合物(II)若しくは化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含有する。
[α-synuclein aggregate binding agent]
In one embodiment, the invention provides alpha-synuclein aggregate binding agents. The α-synuclein aggregate binding agent (hereinafter also referred to as a binding agent) of this embodiment is compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof. contains.
 化合物(I)、化合物(II)及び化合物(III)は、タウタンパク質及びアミロイドβの凝集体よりもαシヌクレイン凝集体への結合選択性が高い。また、同様に、化合物(I)、化合物(II)及び化合物(III)の医薬として許容し得る塩、並びに化合物(I)、化合物(II)及び化合物(III)の溶媒和物も、タウタンパク質及びアミロイドβの凝集体よりもαシヌクレイン凝集体への結合選択性が高い。また、化合物(I)、化合物(II)及び化合物(III)は、蛍光を発する。また、上述の通り、化合物(I)、化合物(II)及び化合物(III)において、1個又はそれ以上の原子を該原子の放射性同位体とすることができる。したがって、本実施形態の結合剤は、脳内に蓄積したαシヌクレイン凝集体の光学イメージング又は放射イメージングのための分子プローブとして用いることができる。 Compound (I), compound (II) and compound (III) have higher binding selectivity to α-synuclein aggregates than to aggregates of tau protein and amyloid β. Similarly, pharmaceutically acceptable salts of Compound (I), Compound (II) and Compound (III), and solvates of Compound (I), Compound (II) and Compound (III) also include tau protein. and more selective binding to α-synuclein aggregates than to aggregates of amyloid β. In addition, compound (I), compound (II) and compound (III) emit fluorescence. Also, as described above, in compound (I), compound (II) and compound (III), one or more atoms can be a radioactive isotope of that atom. Therefore, the binding agents of this embodiment can be used as molecular probes for optical or radioimaging of α-synuclein aggregates accumulated in the brain.
 αシヌクレイン凝集体結合剤は、医薬として許容し得る担体を含むことができる。該医薬として許容し得る担体の例を挙げると、水、食塩水、生理食塩水又はリン酸緩衝食塩水(PBS)、塩化ナトリウム注射液、リンゲル注射液、等張性デキストロース注射液、無菌水注射液、デキストロース、及び乳酸リンゲル注射液などがある。 The α-synuclein aggregate binding agent can contain a pharmaceutically acceptable carrier. Examples of such pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
 αシヌクレイン凝集体結合剤に含まれる化合物(I)、化合物(II)又は化合物(III)、その医薬として許容される塩、又はその溶媒和物、及び医薬として許容し得る担体の含有量は、特に制限はない。これらの含有量は、使用される化合物の種類;投与される哺乳動物の年齢、体重、健康状態、性別及び食事内容;投与の回数、及び投与経路;治療期間;同時に使用される他の薬剤など、様々な要因により決定される。医薬として許容し得る担体の含有量は、αシヌクレイン凝集体結合剤の1~99重量%の量とすることができる。αシヌクレイン凝集体結合剤は、化合物(I)、化合物(II)又は化合物(III)を、例えば、対象の体重(kg)あたりの化合物量が、5ng/kg~5mg/kgの量で投与できるように調製される。好ましくは、当該化合物量の下限は5ng/kg以上、0.01mg/kg以上、0.05mg/kg以上、又は、0.1mg/kg以上である。また、当該化合物量の上限は5mg/kg以下、3mg/kg以下、1mg/kg以下、又は、20μg/kg以下の量である。 The content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the α-synuclein aggregate-binding agent is There are no particular restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors. The content of pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the alpha-synuclein aggregate binding agent. The α-synuclein aggregate binding agent can be administered with compound (I), compound (II) or compound (III), for example, in an amount of 5 ng/kg to 5 mg/kg of the compound per body weight (kg) of the subject. prepared as follows. Preferably, the lower limit of the amount of the compound is 5 ng/kg or more, 0.01 mg/kg or more, 0.05 mg/kg or more, or 0.1 mg/kg or more. The upper limit of the amount of the compound is 5 mg/kg or less, 3 mg/kg or less, 1 mg/kg or less, or 20 μg/kg or less.
 [αシヌクレイン凝集体の光学イメージング用組成物]
 一実施形態において、本発明は、αシヌクレイン凝集体の光学イメージング用組成物を提供する。本実施形態のαシヌクレイン凝集体の光学イメージング用組成物(以下、単に、「光学イメージング用組成物」とも記載する)は、上述の本実施形態に係る結合剤を含む。当該光学イメージングは、インビトロ、エキソビボ、及びインビボイメージングを含む。
[Composition for optical imaging of α-synuclein aggregates]
In one embodiment, the invention provides compositions for optical imaging of alpha-synuclein aggregates. The composition for optical imaging of α-synuclein aggregates of the present embodiment (hereinafter also simply referred to as the “composition for optical imaging”) contains the above-described binder according to the present embodiment. Such optical imaging includes in vitro, ex vivo, and in vivo imaging.
 光学イメージングとしては、例えば、蛍光顕微鏡測定法、多光子イメージング法、二光子イメージング法、及び近赤外蛍光イメージング法が挙げられる。 Optical imaging includes, for example, fluorescence microscopy, multiphoton imaging, two-photon imaging, and near-infrared fluorescence imaging.
 光学イメージング用組成物は、医薬として許容し得る担体を含むことができる。該医薬として許容し得る担体の例を挙げると、水、食塩水、生理食塩水又はリン酸緩衝食塩水(PBS)、塩化ナトリウム注射液、リンゲル注射液、等張性デキストロース注射液、無菌水注射液、デキストロース、及び乳酸リンゲル注射液などがある。 The optical imaging composition can contain a pharmaceutically acceptable carrier. Examples of such pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
 光学イメージング用組成物に含まれる化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物、及び医薬として許容し得る担体の含有量は、特に制限はない。これらの含有量は、使用される化合物の種類;投与される哺乳動物の年齢、体重、健康状態、性別及び食事内容;投与の回数、及び投与経路;治療期間;同時に使用される他の薬剤など、様々な要因により決定される。医薬として許容し得る担体の含有量は、光学イメージング用組成物の1~99重量%の量とすることができる。光学イメージング用組成物は、化合物(I)、化合物(II)又は化合物(III)を、例えば、対象の体重(kg)あたりの化合物量(mg)が、0.01mg/kg~5mg/kg、好ましくは0.05mg/kg~3mg/kg、さらに好ましくは、0.1mg/kg~1mg/kgの量で投与できるように調製される。 The content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the optical imaging composition is There are no restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors. The content of the pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the optical imaging composition. The composition for optical imaging contains compound (I), compound (II) or compound (III), for example, the compound amount (mg) per subject body weight (kg) is 0.01 mg/kg to 5 mg/kg, It is prepared so that it can be administered in an amount of preferably 0.05 mg/kg to 3 mg/kg, more preferably 0.1 mg/kg to 1 mg/kg.
 [αシヌクレイン凝集体の放射イメージング用組成物]
 一実施形態において、本発明は、αシヌクレイン凝集体の放射イメージング用組成物を提供する。本実施形態のαシヌクレイン凝集体の照射イメージング用組成物(以下、単に、「放射イメージング用組成物」とも記載する)は、化合物(I)、化合物(II)及び化合物(III)における1個又はそれ以上の原子が該原子の放射性同位体である化合物を含有する、上述の本実施形態に係る結合剤を含む。当該放射イメージングは、インビトロ、エキソビボ、及びインビボイメージングを含む。
[Composition for radiation imaging of α-synuclein aggregates]
In one embodiment, the invention provides a composition for radioimaging alpha-synuclein aggregates. The composition for radiation imaging of α-synuclein aggregates of the present embodiment (hereinafter also simply referred to as "composition for radiation imaging") is compound (I), compound (II) and compound (III), or Includes binders according to this embodiment described above that contain compounds in which a further atom is a radioactive isotope of that atom. Such radioimaging includes in vitro, ex vivo, and in vivo imaging.
 放射イメージングとしては、例えば、ポジトロン断層撮影法(Positron Emission Tomography、PET)、単一光子放射断層撮影法(Single photon emission computed tomography、SPECT)、及びオートラジオグラフィが挙げられる。 Radiation imaging includes, for example, positron emission tomography (PET), single photon emission computed tomography (SPECT), and autoradiography.
 放射イメージング用組成物は、医薬として許容し得る担体を含むことができる。該医薬として許容し得る担体の例を挙げると、水、食塩水、生理食塩水又はリン酸緩衝食塩水(PBS)、塩化ナトリウム注射液、リンゲル注射液、等張性デキストロース注射液、無菌水注射液、デキストロース、及び乳酸リンゲル注射液などがある。 The radioimaging composition can contain a pharmaceutically acceptable carrier. Examples of such pharmaceutically acceptable carriers include water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringer's injection, isotonic dextrose injection, sterile water injection. liquid, dextrose, and lactated Ringer's injection.
 放射イメージング用組成物に含まれる化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物、及び医薬として許容し得る担体の含有量は、特に制限はない。これらの含有量は、使用される化合物の種類;投与される哺乳動物の年齢、体重、健康状態、性別及び食事内容;投与の回数、及び投与経路;治療期間;同時に使用される他の薬剤など、様々な要因により決定される。医薬として許容し得る担体の含有量は、放射イメージング用組成物の1~99重量%の量とすることができる。放射イメージング用組成物は、化合物(I)、化合物(II)又は化合物(III)を、例えば、対象の体重(kg)あたりの化合物量が、5ng/kg~5mg/kg、好ましくは5ng/kg~20μg/kgの量で投与できるように調製される。 The content of compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt or solvate thereof, and a pharmaceutically acceptable carrier contained in the radioimaging composition is particularly There are no restrictions. These contents include the type of compound used; the age, weight, health condition, sex and diet content of the mammal to be administered; the number of administrations and administration route; the duration of treatment; , determined by a variety of factors. The content of the pharmaceutically acceptable carrier can be in an amount of 1-99% by weight of the radioimaging composition. The radiation imaging composition contains compound (I), compound (II) or compound (III), for example, in an amount of the compound per body weight (kg) of the subject of 5 ng/kg to 5 mg/kg, preferably 5 ng/kg. It is formulated to be dosed at ˜20 μg/kg.
 [αシヌクレイン凝集体が関連する疾患の診断薬、又は当該疾患の治療又は予防のためのコンパニオン診断薬]
 一実施形態において、本発明は、αシヌクレイン凝集体が関連する疾患の診断薬、又は当該疾患の治療又は予防のためのコンパニオン診断薬を提供する。本実施形態のαシヌクレイン凝集体が関連する疾患の診断薬、又は当該疾患の治療又は予防のためのコンパニオン診断薬(以下、コンパニオン診断薬ともいう)は、上述の本実施形態に係る結合剤を含む。治療のためのコンパニオン診断薬とは、疾患であることが判明した場合に、治療が見込めるかどうかを判断するための診断薬である。また、予防のためのコンパニオン診断薬とは、疾患の前駆状態であることが判明した場合に、今後の発症を予測する、若しくは発症を抑制する予防が見込めるかどうかを判断するための診断薬である。
[Diagnostic agent for diseases associated with α-synuclein aggregates, or companion diagnostic agent for treatment or prevention of the disease]
In one embodiment, the present invention provides diagnostics for diseases associated with α-synuclein aggregates, or companion diagnostics for the treatment or prevention of such diseases. The diagnostic agent for diseases associated with α-synuclein aggregates of the present embodiment, or the companion diagnostic agent for treating or preventing the disease (hereinafter also referred to as the companion diagnostic agent) is the binding agent according to the present embodiment described above. include. A companion diagnostic for treatment is a diagnostic that, if found to be diseased, is used to determine whether treatment is likely. In addition, companion diagnostics for prevention are diagnostics for predicting future onset or judging whether prevention can be expected to suppress onset when it turns out to be a prodromal state of a disease. be.
 本実施形態に係る診断薬を用いて、対象から得られる脳内αシヌクレイン凝集体の量及び/又は分布量に関するデータを、予め取得した疾患とαシヌクレイン凝集体の量及び/又は分布量との相関性に照合することで、対象の疾患に関する診断(具体的には、疾患の罹患有無、重篤性、及び発作可能性等)が可能である。 Using the diagnostic agent according to the present embodiment, data on the amount and/or distribution of brain α-synuclein aggregates obtained from a subject are compared with the previously obtained disease and the amount and/or distribution of α-synuclein aggregates. By checking the correlation, it is possible to diagnose the target disease (specifically, the presence or absence of the disease, the severity, the possibility of seizure, etc.).
 また、コンパニオン診断薬を用いて、対象から得られる脳内αシヌクレイン凝集体の量及び/又は分布量に関するデータを、予め取得した疾患と脳内αシヌクレイン凝集体の量及び/又は分布量との相関性に照合することで、対象の疾患状態を把握することができる。そのため、これに基づき、疾患の予防/治療計画(投与する予防薬又は治療薬の種類、組み合わせ、用量、及び用途など)を策定することができる。 In addition, using a companion diagnostic agent, data on the amount and/or distribution of brain α-synuclein aggregates obtained from a subject can be obtained by comparing the previously obtained disease with the amount and/or distribution of brain α-synuclein aggregates. By checking the correlation, the disease state of the subject can be grasped. Therefore, based on this, disease prophylaxis/treatment regimens (types of prophylactic or therapeutic agents to be administered, combinations, doses, uses, etc.) can be formulated.
 本発明の一実施形態は、αシヌクレイン凝集体が関連する疾患の治療又は予防のための医薬であって、コンパニオン診断で得られる脳内αシヌクレイン凝集体の量及び/又は分布に関するデータに基づく投与計画で投与される医薬にも関する。 One embodiment of the present invention is a medicament for the treatment or prevention of diseases associated with α-synuclein aggregates, which is administered based on data on the amount and/or distribution of brain α-synuclein aggregates obtained by companion diagnostics. It also relates to medicaments administered on a schedule.
 [脳内に蓄積する物質が関連する疾患の診断キット]
 本発明の脳内に蓄積する物質が関連する疾患の診断キット(以下、診断キットともいう)は、上述の本実施形態に係る結合剤を含む。
[Diagnostic kit for diseases associated with substances accumulated in the brain]
A diagnostic kit for a disease associated with a substance that accumulates in the brain of the present invention (hereinafter also referred to as a diagnostic kit) includes the above-described binding agent according to this embodiment.
 脳内に蓄積する物質としては、少なくともαシヌクレイン凝集体を含み、その他に、タウタンパク質又はアミロイドβの凝集体が挙げられる。 Substances that accumulate in the brain include at least α-synuclein aggregates, and also include tau protein or amyloid β aggregates.
 脳内に蓄積する物質が関連する疾患としては、少なくともαシヌクレイン凝集体が関連する疾患を含み、当該疾患としては、パーキンソン病、レビー小体型認知症(DLB)、及び多系統萎縮症(MSA)が挙げられる。脳内に蓄積する物質が関連する疾患は、この他に、タウタンパク質又はアミロイドβの凝集体が関連する疾患であるアルツハイマー病(AD)及び前頭側頭葉変性症が挙げられる。 Diseases associated with substances that accumulate in the brain include at least diseases associated with α-synuclein aggregates, such as Parkinson's disease, dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). is mentioned. Other diseases associated with substances that accumulate in the brain include Alzheimer's disease (AD) and frontotemporal lobar degeneration, which are diseases associated with aggregates of tau protein or amyloid β.
 本実施形態の結合剤はタウタンパク質及びアミロイドβの凝集体よりもαシヌクレイン凝集体への結合選択性が高い。一方で、例えば、特許文献1記載の化合物は、αシヌクレイン凝集体よりもタウタンパク質凝集体への結合性が高いことが、本発明者らの検討の結果から判明した。 The binding agent of the present embodiment has higher binding selectivity to α-synuclein aggregates than to aggregates of tau protein and amyloid β. On the other hand, the results of studies by the present inventors revealed that, for example, the compounds described in Patent Document 1 have higher binding properties to tau protein aggregates than to α-synuclein aggregates.
 本実施形態の一実施態様において、診断キットは、αシヌクレイン凝集体への結合選択性が高い化合物(I)、化合物(II)若しくは化合物(III)又はその医薬として許容し得る塩若しくはその溶媒和物、及びタウタンパク質凝集体への結合選択性が高い他の化合物(例えば、特許文献1記載の化合物)の両方を含むことができる。このような診断キットにおいては、前者の検出結果(検出される光若しくは放射線の量及び/又は分布)と後者の検出結果とを比較することで、イメージングされた各領域に存在する物質がどの物質なのか特定できる。具体的にはαシヌクレイン凝集体と、タウタンパク質凝集体とを分類することができ、さらに各々の存在量を定量することもできる。このため、αシヌクレイン凝集体が関連する疾患、及び/又は、タウタンパク質が関連する疾患を高精度に診断することができる。例えば、αシヌクレイン凝集体への結合性と、タウタンパク質凝集体への結合性との比を、本実施形態の結合剤と、タウタンパク質凝集体への結合選択性が高い物質(例えば、特許文献1記載の化合物)との各々について予め特定しておく。そのうえで、被検体での前者及び後者投与後の脳内各領域での光又は放射線の量比を測定すれば、予め特定した比と測定した量比との関係から当該領域にαシヌクレイン凝集体、タウタンパク質凝集体のいずれが存在するのかを分類し、各量の定量が可能である。 In one embodiment of this embodiment, the diagnostic kit comprises compound (I), compound (II) or compound (III) with high binding selectivity to α-synuclein aggregates or a pharmaceutically acceptable salt or solvate thereof and other compounds with high binding selectivity to tau protein aggregates (for example, compounds described in Patent Document 1). In such a diagnostic kit, by comparing the former detection result (amount and/or distribution of detected light or radiation) and the latter detection result, it is possible to determine which substance is present in each region imaged. can be identified. Specifically, α-synuclein aggregates and tau protein aggregates can be classified, and the abundance of each can be quantified. Therefore, diseases associated with α-synuclein aggregates and/or diseases associated with tau protein can be diagnosed with high accuracy. For example, the binding agent of the present embodiment and a substance with high binding selectivity to tau protein aggregates (e.g., patent literature 1) are specified in advance. Then, by measuring the amount ratio of light or radiation in each area of the brain after administration of the former and the latter in the subject, α-synuclein aggregates, α-synuclein aggregates, It is possible to classify which tau protein aggregates are present and quantify the amount of each.
 また、本実施形態の上記診断キットを、さらにアミロイドβのイメージング剤と組み合わせた診断キットとすることもできる。アミロイドβのイメージング剤をさらに組み合わせた診断キットは、イメージングされた各領域に存在する物質がどの物質なのか特定できる。具体的にはαシヌクレイン凝集体と、タウタンパク質凝集体と、アミロイドβ凝集体とを分類することができ、さらに各々の存在量を定量することもできる。このため、αシヌクレイン凝集体が関連する疾患、タウタンパク質が関連する疾患、及び/又はアミロイドβが案連する疾患を高精度に診断することができる。 In addition, the diagnostic kit of the present embodiment can be further combined with an amyloid β imaging agent to form a diagnostic kit. A diagnostic kit further combined with an amyloid β imaging agent can specify which substance is present in each region imaged. Specifically, α-synuclein aggregates, tau protein aggregates, and amyloid β aggregates can be classified, and the abundance of each can be quantified. Therefore, diseases associated with α-synuclein aggregates, diseases associated with tau protein, and/or diseases associated with amyloid β can be diagnosed with high accuracy.
 本発明の一実施形態は、αシヌクレイン凝集体が関連する疾患の治療又は予防のための医薬であって、本実施形態に係る診断キットで得られるαシヌクレイン凝集体を含む脳内蓄積物質の量及び/又は分布に関するデータに基づく投与計画で投与される医薬にも関する。 One embodiment of the present invention is a medicament for treating or preventing diseases associated with α-synuclein aggregates, wherein the amount of brain accumulated substances containing α-synuclein aggregates obtained from the diagnostic kit according to this embodiment is and/or a medicament administered in a dosing regimen based on distribution data.
 [光学イメージング方法]
 一実施形態において、本発明は、光学イメージング方法を提供する。本実施形態の光学イメージング方法は、上述の本実施形態に係る結合剤を投与された被検体の生体脳に脳外から第1の波長の光を照射した後に、該脳から発せられる、第1の波長とは異なる第2の波長の光を検出する工程を含む。
[Optical imaging method]
In one embodiment, the invention provides an optical imaging method. In the optical imaging method of the present embodiment, after the brain of a living subject to whom the binding agent of the present embodiment has been administered is irradiated with light of a first wavelength from outside the brain, the first detecting light of a second wavelength different from the wavelength of the.
 被検体に有効量の結合剤を投与すると、生体脳に移行した結合剤が、生体脳内に存在するαシヌクレイン凝集体に結合する。結合剤を励起させる第1の波長の光を、結合剤を投与された被検体の生体脳に脳外から照射し、脳内の結合剤から発せられる第2の波長の光(例えば、蛍光)を検出することにより、αシヌクレイン凝集体の光学イメージング(画像化)をすることができる。 When an effective amount of binding agent is administered to a subject, the binding agent transferred to the living brain binds to α-synuclein aggregates present in the living brain. Light of a first wavelength that excites the binding agent is irradiated extracerebrally into the living brain of a subject administered the binding agent, and light of a second wavelength (e.g., fluorescence) emitted from the binding agent in the brain. can be used for optical imaging of α-synuclein aggregates.
 被検体としては、哺乳動物が挙げられる。哺乳動物は、例えば、ヒト、ラット、マウス、ウサギ、モルモット、ハムスター、サル、イヌ、フェレット、又はミニブタなどを含む。好ましくは、哺乳動物は、ヒトである。  Subjects include mammals. Mammals include, for example, humans, rats, mice, rabbits, guinea pigs, hamsters, monkeys, dogs, ferrets, minipigs, and the like. Preferably, the mammal is human.
 投与方法は、特に制限されないが、例えば、経口投与、及び静脈内投与又は腹腔内投与等の非経口投与がある。好ましくは、静脈内投与又は腹腔内投与である。最も好ましくは、静脈内投与である。投与量は、対象の体重(kg)あたりの化合物(I)、化合物(II)又は化合物(III)の量(mg)が、0.01mg/kg~5mg/kg、0.05mg/kg~3mg/kg、又は0.1mg/kg~1mg/kgであることが好ましく、最も好ましくは、0.1mg/kg~1mg/kgである。 The administration method is not particularly limited, but includes, for example, oral administration and parenteral administration such as intravenous administration or intraperitoneal administration. Intravenous administration or intraperitoneal administration is preferred. Most preferred is intravenous administration. The dosage is such that the amount (mg) of compound (I), compound (II) or compound (III) per body weight (kg) of the subject is 0.01 mg/kg to 5 mg/kg, 0.05 mg/kg to 3 mg. /kg, or 0.1 mg/kg to 1 mg/kg, most preferably 0.1 mg/kg to 1 mg/kg.
 [放射イメージング方法]
 一実施形態において、本発明は、放射イメージング方法を提供する。本実施形態の放射イメージング方法は、1個又はそれ以上の原子が該原子の放射性同位体である化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含む本実施形態に係る結合剤を投与された被検体の生体脳から発せられる放射線を検出する工程を含む。
[Radiation Imaging Method]
In one embodiment, the invention provides a radiation imaging method. The radioimaging method of this embodiment comprises compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a Detecting radiation emitted from the living brain of a subject administered a binding agent according to the present embodiments, including the solvate.
 被検体に有効量の結合剤を投与すると、生体脳に移行した結合剤が、生体脳内に存在するαシヌクレイン凝集体に結合する。脳内の結合剤から発せられる放射線を検出することにより、αシヌクレイン凝集体の放射イメージング(画像化)をすることができる。 When an effective amount of binding agent is administered to a subject, the binding agent transferred to the living brain binds to α-synuclein aggregates present in the living brain. Radiation imaging of α-synuclein aggregates can be performed by detecting radiation emitted from the binding agent in the brain.
 被検体としては、哺乳動物が挙げられる。哺乳動物は、例えば、ヒト、ラット、マウス、ウサギ、モルモット、ハムスター、サル、イヌ、フェレット、又はミニブタなどを含む。好ましくは、哺乳動物は、ヒトである。  Subjects include mammals. Mammals include, for example, humans, rats, mice, rabbits, guinea pigs, hamsters, monkeys, dogs, ferrets, minipigs, and the like. Preferably, the mammal is human.
 投与方法は、特に制限されないが、例えば、経口投与、及び静脈内投与又は腹腔内投与等の非経口投与がある。好ましくは、静脈内投与又は腹腔内投与である。最も好ましくは、静脈内投与である。投与量は、対象の体重(kg)あたりの化合物(I)、化合物(II)又は化合物(III)の量(mg)が、5ng/kg~5mg/kgであることが好ましく、5ng~20μg/kgであることがより好ましい。投与放射能量は、個体あたり37MBq~7.4GBqであることが好ましく、370MBq~3,700MBqであることがより好ましい。 The administration method is not particularly limited, but includes, for example, oral administration and parenteral administration such as intravenous administration or intraperitoneal administration. Intravenous administration or intraperitoneal administration is preferred. Most preferred is intravenous administration. The dosage is preferably 5 ng/kg to 5 mg/kg of compound (I), compound (II) or compound (III) per body weight (kg) of the subject, and 5 ng to 20 μg/kg. kg is more preferred. The amount of radioactivity administered is preferably 37 MBq to 7.4 GBq, more preferably 370 MBq to 3,700 MBq per individual.
 [脳内αシヌクレイン凝集体が関連する疾患の治療薬又は予防薬のスクリーニング方法]
 一実施形態において、本発明は、脳内αシヌクレイン凝集体が関連する疾患の治療又は予防薬のスクリーニング方法を提供する。本実施形態の脳内αシヌクレイン凝集体が関連する疾患の治療又は予防薬のスクリーニング方法(以下、スクリーニング方法ともいう)は、被検体への候補物質の投与前後における、本実施形態に係る光学イメージング方法又は放射イメージング方法によって検出される光又は放射線の量及び/又は分布の差異に基づいて、候補物質を選抜する工程を有する。
[Screening method for therapeutic or preventive drug for diseases associated with brain α-synuclein aggregates]
In one embodiment, the present invention provides a method of screening for therapeutic or preventive agents for diseases associated with brain α-synuclein aggregates. The screening method for therapeutic or preventive drugs for diseases associated with brain α-synuclein aggregates of the present embodiment (hereinafter also referred to as screening method) comprises optical imaging according to the present embodiment before and after administration of a candidate substance to a subject. selecting candidate substances based on differences in the amount and/or distribution of light or radiation detected by the method or radiation imaging method.
 脳内αシヌクレイン凝集体が関連する疾患は、上記[脳内に蓄積する物質が関連する疾患の診断キット]に記載された少なくともαシヌクレイン凝集体が関連する疾患と同様である。 Diseases associated with brain α-synuclein aggregates are at least the same as the diseases associated with α-synuclein aggregates described in the above [Diagnostic kit for diseases associated with substances that accumulate in the brain].
 また、被検体及び投与方法は、上記[光学イメージング方法]及び[放射イメージング方法]に記載されたものと同様である。 In addition, the subject and administration method are the same as those described in [Optical Imaging Method] and [Radiation Imaging Method] above.
 例えば、候補物質の投与後、結合剤の蛍光等の光又は放射線の量(強度)が、候補物質を投与する前のものと比較して減少している場合、候補物質が、該疾患又は症状の治療用化合物として有用であり得る。 For example, if the amount (intensity) of light or radiation, such as fluorescence of the binding agent, decreases after administration of the candidate substance compared to before administration of the candidate substance, the candidate substance is associated with the disease or condition. can be useful as a therapeutic compound for
 また、被検体において検出される光又は放射線の量及び/又は分布を、他の正常な哺乳動物のものと比較し、候補化合物の投与後に投与前よりも正常な哺乳動物のものに近づく場合、該候補化合物が、該疾患又は症状の治療用化合物として有用であり得る。 Also, if the amount and/or distribution of light or radiation detected in a subject is compared to that of other normal mammals and approaches that of a normal mammal after administration of the candidate compound than before administration, Said candidate compound may be useful as a therapeutic compound for said disease or condition.
 例えば、候補物質を投与された対象から得られる候補物質投与前後の結合剤の蛍光等の光又は放射線の量(強度)及び/又は分布に関するデータを、予め取得した候補物質を投与されていない哺乳動物における、脳内αシヌクレイン凝集体が関連する疾患発症前後での結合剤の蛍光等の光又は放射線の増加量及び/又は分布の変化と比較する。そして、疾患発症後に見られる当該蛍光等の光又は放射線量の増加が、候補物質の投与により抑制されている、及び/又は、当該蛍光等の光又は放射線量が、候補物質の投与後において、投与前と同様に正常な哺乳動物のものと近い値を示す場合、候補物質が、該疾患又は症状の予防用化合物として有用であり得る。同様に、疾患発症後に見られる当該蛍光等の光又は放射線の分布の変化が、候補物質の投与により抑制されるか、及び/又は、当該蛍光等の光又は放射線の分布の変化が、候補物質の投与後において、投与前と同様に正常な哺乳動物における分布に近い場合にも、候補物質が、該疾患又は症状の予防用化合物として有用であり得る。 For example, data on the amount (intensity) and/or distribution of light or radiation such as fluorescence of a binding agent before and after administration of a candidate substance obtained from a subject to whom the candidate substance has been administered may be collected from a previously acquired mammal not administered the candidate substance. The increase and/or change in distribution of light or radiation such as fluorescence of the binding agent before and after the onset of disease involving brain α-synuclein aggregates in animals is compared. Then, an increase in the amount of light such as fluorescence or radiation seen after the onset of the disease is suppressed by administration of the candidate substance, and/or the amount of light such as fluorescence or radiation is reduced after administration of the candidate substance, A candidate substance may be useful as a preventive compound for the disease or condition if it exhibits values close to those of normal mammals as before administration. Similarly, the change in the distribution of light such as fluorescence or radiation seen after the onset of the disease is suppressed by administration of the candidate substance, and/or the change in the distribution of light such as fluorescence or radiation is suppressed by the candidate substance. A candidate substance may also be useful as a compound for prophylaxis of the disease or condition if, after administration of the drug, the distribution in a normal mammal is as close to that as before administration.
 [脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法]
 一実施形態において、本発明は、脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法を提供する。本実施形態の脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法は、化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含む上記結合剤を投与された被検体の生体脳に脳外から第1の波長の光を照射した後に、該脳から発せられる、第1の波長とは異なる第2の波長の光を検出する工程を含み、該検出した光の量及び/又は分布に基づいて、脳内αシヌクレイン凝集体の蓄積を定量又は判定する。この方法は、光学イメージングにより、脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法である。
[Method for Quantifying or Determining Accumulation of Brain α-Synuclein Aggregates]
In one embodiment, the invention provides a method of quantifying or determining the accumulation of brain α-synuclein aggregates. The method of quantifying or determining the accumulation of brain α-synuclein aggregates of this embodiment includes compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof. A step of detecting light of a second wavelength different from the first wavelength emitted from the brain after irradiating the living brain of the subject to which the binding agent has been administered from outside the brain with the light of the first wavelength. and quantifying or determining the accumulation of brain α-synuclein aggregates based on the amount and/or distribution of the detected light. This method is a method for quantifying or determining the accumulation of brain α-synuclein aggregates by optical imaging.
 被検体及び投与方法は、上記[光学イメージング方法]に記載されたものと同様である。 The subject and administration method are the same as those described in [Optical Imaging Method] above.
 被検体において検出される光の量及び/又は分布と、他の正常な哺乳動物のものとの差を求めることで、脳内のαシヌクレイン凝集体の蓄積を定量すること、及び脳内のαシヌクレイン凝集体の蓄積の有無を判定することができる。 quantifying the accumulation of alpha-synuclein aggregates in the brain by determining the difference between the amount and/or distribution of light detected in a subject and that of other normal mammals; The presence or absence of accumulation of synuclein aggregates can be determined.
 本実施形態の脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法の別の態様では、1個又はそれ以上の原子が該原子の放射性同位体である化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含む本実施形態に係る結合剤を投与された被検体の生体脳から発せられる放射線を検出する工程を含み、該検出した放射線の量及び/又は分布に基づいて、脳内αシヌクレイン凝集体の蓄積を定量又は判定する。この方法は、放射イメージングにより、脳内αシヌクレイン凝集体の蓄積を定量又は判定する方法である。 In another aspect of the method of quantifying or determining the accumulation of brain α-synuclein aggregates of this embodiment, compound (I), compound (II), or detecting radiation emitted from the living brain of a subject administered a binding agent according to this embodiment comprising compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof; Accumulation of brain α-synuclein aggregates is quantified or determined based on the amount and/or distribution of radiation. This method is a method for quantifying or determining the accumulation of α-synuclein aggregates in the brain by radioimaging.
 被検体及び投与方法は、上記[放射イメージング方法]に記載されたものと同様である。 The subject and administration method are the same as those described in [Radiation Imaging Method] above.
 被検体において検出される放射線の量及び/又は分布と、他の正常な哺乳動物のものとの差を求めることで、脳内のαシヌクレイン凝集体の蓄積を定量すること、及び脳内のαシヌクレイン凝集体の蓄積の有無を判定することができる。 quantifying the accumulation of alpha-synuclein aggregates in the brain by determining the difference between the amount and/or distribution of radiation detected in a subject and that of other normal mammals; The presence or absence of accumulation of synuclein aggregates can be determined.
 [脳内に蓄積する物質の分類及び蓄積を判定する方法]
 一実施形態において、本発明は、脳内に蓄積する物質の分類及び蓄積を判定する方法を提供する。本発明の脳内に蓄積する物質の分類及び蓄積を判定する方法は、化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含む本実施形態に係る結合剤を投与された被検体の生体脳に脳外から第1の波長の光を照射した後に、該脳から発せられる、第1の波長とは異なる第2の波長の光を検出する第1の工程と、タウタンパク質凝集体への結合選択性が高い物質(例えば、特許文献1記載の化合物)を、第1の工程と異なる時期に投与された該被検体の生体脳に脳外から第3の波長の光を照射した後に、該脳から発せられる、第3の波長とは異なる第4の波長の光を検出する第2の工程と、を含み、第1の工程において検出した光の量及び/又は分布データと、第2の工程において検出した光の量及び/又は分布データと、に基づいて、脳内に蓄積する物質の分類及び蓄積を判定する。この方法は、光学イメージングにより、脳内に蓄積する物質の分類及び蓄積を判定する方法である。
[Method for determining classification and accumulation of substances accumulated in the brain]
In one embodiment, the invention provides a method for determining the classification and accumulation of substances that accumulate in the brain. The method for determining the classification and accumulation of substances that accumulate in the brain of the present invention includes compound (I), compound (II) or compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof. After irradiating the living brain of a subject to whom the binding agent according to the embodiment has been administered with light of a first wavelength from outside the brain, light of a second wavelength different from the first wavelength emitted from the brain is irradiated. In the first step of detecting, a substance with high binding selectivity to tau protein aggregates (for example, the compound described in Patent Document 1) is administered to the living brain of the subject at a different time from the first step. a second step of detecting light of a fourth wavelength different from the third wavelength emitted from the brain after irradiating the light of the third wavelength from outside the brain; Based on the amount and/or distribution data of light detected and the amount and/or distribution data of light detected in the second step, the classification and accumulation of substances accumulating in the brain are determined. This method uses optical imaging to determine the classification and accumulation of substances that accumulate in the brain.
 被検体及び投与方法は、上記[光学イメージング方法]に記載されたものと同様である。 The subject and administration method are the same as those described in [Optical Imaging Method] above.
 αシヌクレイン凝集体への結合選択性が高い結合剤の投与に起因する脳から発せられる光を検出する第1の工程と、同じ被検体における、タウタンパク質凝集体への結合選択性が高い物質の投与に起因する脳から発せられる光を検出する第2の工程との両方を含むため、第1の工程の検出結果(検出される光の量及び/又は分布)と第2の工程の検出結果とを比較することで、脳内に蓄積する物質がαシヌクレイン凝集体及び/又はタウタンパク質凝集体であるか否かの分類及び各凝集体の蓄積を判定することができる。 A first step of detecting light emitted from the brain resulting from administration of a binding agent with high binding selectivity to alpha-synuclein aggregates, and a binding agent with high binding selectivity to tau protein aggregates in the same subject. and a second step of detecting light emitted from the brain resulting from administration, the detection result of the first step (amount and/or distribution of detected light) and the detection result of the second step By comparing , it is possible to classify whether substances that accumulate in the brain are α-synuclein aggregates and/or tau protein aggregates, and determine the accumulation of each aggregate.
 本実施形態の脳内に蓄積する物質の分類及び蓄積を判定する方法の別の態様では、1個又はそれ以上の原子が該原子の放射性同位体である化合物(I)、化合物(II)又は化合物(III)、その医薬として許容し得る塩、又はその溶媒和物を含む本実施形態に係る結合剤を投与された被検体の生体脳から発せられる放射線を検出する第1の工程と、タウタンパク質凝集体への結合選択性が高い放射性の物質(例えば、特許文献1記載の化合物)を、第1の工程と異なる時期に投与された該被検体の脳から発せられる放射線を検出する第2の工程と、を含み、第1の工程において検出した放射線の量及び/又は分布データと、第2の工程において検出した放射線の量及び/又は分布データと、に基づいて、脳内に蓄積する物質の分類及び蓄積を判定する。この方法は、放射イメージングにより、脳内に蓄積する物質の分類及び蓄積を判定する方法である。 In another aspect of the method for determining the classification and accumulation of substances that accumulate in the brain of this embodiment, compound (I), compound (II), or a first step of detecting radiation emitted from the living brain of a subject to which a binding agent according to this embodiment comprising compound (III), a pharmaceutically acceptable salt thereof, or a solvate thereof; A second step of detecting radiation emitted from the brain of the subject administered a radioactive substance with high binding selectivity to protein aggregates (for example, the compound described in Patent Document 1) at a time different from that of the first step. Accumulating in the brain based on the amount and/or distribution data of radiation detected in the first step and the amount and/or distribution data of radiation detected in the second step Determine the classification and accumulation of substances. This method is a method of determining the classification and accumulation of substances that accumulate in the brain by radiation imaging.
 被検体及び投与方法は、上記[放射イメージング方法]に記載されたものと同様である。 The subject and administration method are the same as those described in [Radiation Imaging Method] above.
 αシヌクレイン凝集体への結合選択性が高い結合剤の投与に起因して脳から発せられる放射線を検出する第1の工程と、同じ被検体における、タウタンパク質凝集体への結合選択性が高い物質の投与に起因する脳から発せられる放射線を検出する第2の工程との両方を含むため、第1の工程の検出結果(検出される放射線の量及び/又は分布)と第2の工程の検出結果とを比較することで、脳内に蓄積する物質がαシヌクレイン凝集体及び/又はタウタンパク質凝集体であるか否かの分類及び各凝集体の蓄積を判定することができる。 A first step of detecting radiation emitted from the brain due to administration of a binding agent with high binding selectivity to α-synuclein aggregates and a substance with high binding selectivity to tau protein aggregates in the same subject Since it includes both a second step of detecting radiation emitted from the brain resulting from the administration of the detection result of the first step (amount and / or distribution of radiation detected) and the detection of the second step By comparing the results, it is possible to determine whether the substances accumulated in the brain are α-synuclein aggregates and/or tau protein aggregates, and the accumulation of each aggregate.
 (まとめ)
 以下に本発明の実施形態をまとめる。
(summary)
Embodiments of the present invention are summarized below.
 本発明の一態様は、下記式(I)、(II)又は(III)で表される化合物、その医薬として許容し得る塩、又はその溶媒和物である。
Figure JPOXMLDOC01-appb-C000023
 本発明の一態様は、前記式(I)、(II)又は(III)で表される化合物において1個又はそれ以上の原子が該原子の放射性同位体である、前記化合物、その医薬として許容し得る塩、又はその溶媒和物である。
One aspect of the present invention is a compound represented by formula (I), (II) or (III) below, or a pharmaceutically acceptable salt or solvate thereof.
Figure JPOXMLDOC01-appb-C000023
One aspect of the present invention is a pharmaceutically acceptable or a solvate thereof.
 本発明の一態様は、前記化合物、その医薬として許容し得る塩、又はその溶媒和物を含有する、αシヌクレイン凝集体結合剤である。 One aspect of the present invention is an α-synuclein aggregate binding agent containing the compound, a pharmaceutically acceptable salt thereof, or a solvate thereof.
 本発明の一態様は、前記αシヌクレイン凝集体結合剤を含む、αシヌクレイン凝集体の光学イメージング用組成物である。 One aspect of the present invention is a composition for optical imaging of α-synuclein aggregates, containing the α-synuclein aggregate-binding agent.
 本発明の一態様は、前記αシヌクレイン凝集体結合剤を含む、αシヌクレイン凝集体の放射イメージング用組成物である。 One aspect of the present invention is a composition for radioimaging of α-synuclein aggregates, which contains the α-synuclein aggregate-binding agent.
 本発明の一態様は、前記αシヌクレイン凝集体結合剤を投与された被検体の生体脳に脳外から第1の波長の光を照射した後に、前記脳から発せられる、第1の波長とは異なる第2の波長の光を検出する工程を含む、脳内αシヌクレイン凝集体の光学イメージング方法である。 In one aspect of the present invention, after the living brain of a subject to whom the α-synuclein aggregate-binding agent is administered is irradiated with light of a first wavelength from outside the brain, the first wavelength emitted from the brain is A method of optical imaging brain α-synuclein aggregates comprising detecting light of a different second wavelength.
 本発明の一態様は、前記αシヌクレイン凝集体結合剤を投与された被検体の生体脳から発せられる放射線を検出する工程を含む、脳内αシヌクレイン凝集体の放射イメージング方法である。 One aspect of the present invention is a radioimaging method for intracerebral α-synuclein aggregates, comprising the step of detecting radiation emitted from the living brain of a subject administered with the α-synuclein aggregate-binding agent.
 本発明の一態様は、下記式(IV)又は(V)で表される、前記化合物を合成するための、中間体である。
Figure JPOXMLDOC01-appb-C000024
 式(IV)及び式(V)中、
 Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基又はメタンスルホニル基であり、
 Rは、テトラヒドロ-2H-ピラン-2-イル基又はメトキシメチル基であり、
 Rは、水素原子、tert-ブトキシカルボニル基又は2,4-ジメトキシベンジル基であり、
 式(IV)中、
 Xは窒素原子(N)又は置換されていない炭素原子(CH)であり、
 Rは、水素原子、tert-ブトキシカルボニル基又は2,4-ジメトキシベンジル基である。
One aspect of the present invention is an intermediate for synthesizing the compound represented by the following formula (IV) or (V).
Figure JPOXMLDOC01-appb-C000024
In formula (IV) and formula (V),
R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group,
R 2 is a tetrahydro-2H-pyran-2-yl group or a methoxymethyl group,
R 4 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group,
In formula (IV),
X is a nitrogen atom (N) or an unsubstituted carbon atom (CH);
R 3 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group.
 本発明の一態様は、下記式(VI)で表される、前記化合物を合成するための、中間体である。
Figure JPOXMLDOC01-appb-C000025
 式(VI)中、
 Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基又はメタンスルホニル基であり、
 R4’は、水素原子又はtert-ブトキシカルボニル基であり、
 Xは窒素原子(N)又は置換されていない炭素原子(CH)である。
One aspect of the present invention is an intermediate for synthesizing the compound represented by the following formula (VI).
Figure JPOXMLDOC01-appb-C000025
In formula (VI),
R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group,
R 4' is a hydrogen atom or a tert-butoxycarbonyl group,
X is a nitrogen atom (N) or an unsubstituted carbon atom (CH).
 本発明は上述した各実施形態に限定されるものではなく、請求項に示した範囲で種々の変更が可能であり、異なる実施形態にそれぞれ開示された技術的手段を適宜組み合わせて得られる実施形態についても本発明の技術的範囲に含まれる。 The present invention is not limited to the above-described embodiments, but can be modified in various ways within the scope of the claims, and can be obtained by appropriately combining technical means disclosed in different embodiments. is also included in the technical scope of the present invention.
 (製造例)
 以下の製造例(参考例及び実施例)中の「室温」は通常約10℃~約35℃を示す。%は特に断らない限り重量%を示す。また各製造例の使用化合物の説明に関し、「参考例(実施例)Xで製造した」は、「参考例(実施例)Xと同様にして製造した」場合も含むことを意図している。
(Manufacturing example)
"Room temperature" in the following Production Examples (Reference Examples and Examples) usually means about 10°C to about 35°C. % indicates % by weight unless otherwise specified. In addition, regarding the explanation of the compound used in each production example, "manufactured in Reference Example (Example) X" is intended to include the case of "manufactured in the same manner as Reference Example (Example) X".
 製造例のカラムクロマトグラフィーにおける溶出は、特に言及しない限り、TLC(Thin Layer Chromatography,薄層クロマトグラフィー)による観察下に行った。TLC観察においては、TLCプレートとしてメルク(Merck)社製の60 F254を用い、展開溶媒として、カラムクロマトグラフィーで溶出溶媒として用いた溶媒を用いた。また、検出にはUV検出器を採用した。 Elution in column chromatography in Production Examples was performed under observation by TLC (Thin Layer Chromatography) unless otherwise specified. In the TLC observation, 60 F254 manufactured by Merck was used as the TLC plate, and the solvent used as the elution solvent in column chromatography was used as the developing solvent. A UV detector was employed for detection.
 H NMRの解析にはACD/SpecManager(商品名)ソフトウエアなどを用い、得られた解析値を記載した。水酸基及びアミノ基などのプロトンピークが非常に緩やかなピークについては記載していないことがある。 ACD/SpecManager (trade name) software or the like was used for 1 H NMR analysis, and the analytical values obtained were described. Peaks with very slow proton peaks, such as hydroxyl groups and amino groups, are sometimes not described.
 以下の実施例においては下記の略号を使用する。
Boc:tert-ブトキシカルボニル基
DMF:N,N-ジメチルホルムアミド
DMSO:ジメチルスルホキシド
DMSO-d:重ジメチルスルホキシド
H NMR:プロトン核磁気共鳴
M:モル濃度
MeOH:メタノール
TBS:tert-ブチルジメチルシリル基
TEA:トリエチルアミン
TFA:トリフルオロ酢酸
THF:テトラヒドロフラン
THP:テトラヒドロピラニル基
TMS:トリメチルシリル基
Ts:パラトルエンスルホニル基。
The following abbreviations are used in the examples below.
Boc: tert-butoxycarbonyl group DMF: N,N-dimethylformamide DMSO: dimethylsulfoxide DMSO-d 6 : heavy dimethylsulfoxide
1 H NMR: proton nuclear magnetic resonance M: molarity MeOH: methanol TBS: tert-butyldimethylsilyl group TEA: triethylamine TFA: trifluoroacetic acid THF: tetrahydrofuran THP: tetrahydropyranyl group TMS: trimethylsilyl group Ts: paratoluenesulfonyl group .
 参考例T001:ジ-tert-ブチル (5-ヨードピラジン-2-イル)-2-イミドジカルボナートの製造
Figure JPOXMLDOC01-appb-C000026
 5-ヨードピラジン-2-アミン(CAS[886860-50-0])(9.80g)のTHF(50ml)の0℃の溶液に、二炭酸ジ-tert-ブチル(22.3g)のTHF(25ml)溶液と4-ジメチルアミノピリジン(1.35g)を加えた。その混合物を0℃から室温で終夜撹拌後、水と酢酸エチルで希釈して、不溶物を濾別したのち、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮し、標題化合物(17.74g)を茶色固体として得た。
H NMR(300MHz,DMSO-d)δ1.40(18H,s),8.65(1H,d,J=1.3Hz),8.90(1H,d,J=1.4Hz)。
Reference Example T001: Preparation of di-tert-butyl (5-iodopyrazin-2-yl)-2-imidodicarbonate
Figure JPOXMLDOC01-appb-C000026
Di-tert-butyl dicarbonate (22.3 g) in THF ( 25 ml) solution and 4-dimethylaminopyridine (1.35 g) were added. The mixture was stirred at 0° C. to room temperature overnight, diluted with water and ethyl acetate, filtered to remove insoluble matter, and then extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure to give the title compound (17.74 g) as a brown solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.40 (18 H, s), 8.65 (1 H, d, J = 1.3 Hz), 8.90 (1 H, d, J = 1.4 Hz).
 参考例T002:tert-ブチル (5-ヨードピラジン-2-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000027
 参考例T001で製造したジ-tert-ブチル (5-ヨードピラジン-2-イル)-2-イミドジカルボナート(17.7g)のMeOH(200ml)溶液に、炭酸カリウム(6.97g)を室温で加えた。その混合物を室温で終夜撹拌後、減圧下に約4分の1に濃縮し、0℃に冷却後、5%クエン酸水溶液で中和後に水で希釈した。析出した固体を濾取して、水で洗浄後に乾燥し、標題化合物(11.4g)を茶色固体として得た。
H NMR(300MHz,DMSO-d)δ1.48(9H,s),8.58(1H,d,J=1.5Hz),8.89(1H,d,J=1.5Hz),10.33(1H,brs)。
Reference Example T002: Production of tert-butyl (5-iodopyrazin-2-yl) carbamate
Figure JPOXMLDOC01-appb-C000027
Potassium carbonate (6.97 g) was added to a solution of di-tert-butyl (5-iodopyrazin-2-yl)-2-imidodicarbonate (17.7 g) prepared in Reference Example T001 in MeOH (200 ml) at room temperature. added with After stirring the mixture at room temperature overnight, it was concentrated under reduced pressure to about one fourth, cooled to 0° C., neutralized with 5% aqueous citric acid and diluted with water. The precipitated solid was collected by filtration, washed with water and dried to give the title compound (11.4 g) as a brown solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.48 (9H, s), 8.58 (1H, d, J = 1.5 Hz), 8.89 (1H, d, J = 1.5 Hz), 10.33 (1H, brs).
 参考例T003:tert-ブチル (5-ヨードピラジン-2-イル)(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000028
 参考例T002で製造したtert-ブチル (5-ヨードピラジン-2-イル)カルバマート(2.00g)及び炭酸セシウム(3.04g)のDMF(15ml)の0℃の溶液に、ヨウ化メチル(0.504ml)を加えた。その混合物を0℃から室温で終夜撹拌後、0℃に冷却して、水で希釈した。析出した固体を濾取して、水で洗浄後に乾燥し、標題化合物(1.95g)をベージュ色固体として得た。
H NMR(300MHz,DMSO-d)δ1.49(9H,s),3.27(3H,s),8.71(1H,d,J=1.5Hz),8.83(1H,d,J=1.5Hz)。
Reference Example T003: Production of tert-butyl (5-iodopyrazin-2-yl) (methyl) carbamate
Figure JPOXMLDOC01-appb-C000028
Methyl iodide (0 .504 ml) was added. The mixture was stirred at 0° C. to room temperature overnight, then cooled to 0° C. and diluted with water. The precipitated solid was collected by filtration, washed with water and dried to give the title compound (1.95 g) as a beige solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.49 (9H, s), 3.27 (3H, s), 8.71 (1H, d, J = 1.5 Hz), 8.83 (1H, d, J=1.5 Hz).
 参考例T004:tert-ブチル メチル(5-((トリメチルシリル)エチニル)ピラジン-2-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000029
 参考例T003で製造したtert-ブチル (5-ヨードピラジン-2-イル)(メチル)カルバマート(1.95g)、エチニルトリメチルシラン(1.62ml)及びジクロロビス(トリフェニルホスフィン)パラジウム(II)(123mg)のTHF(6ml)とTEA(6ml)との混合液に、ヨウ化銅(I)(66mg)を窒素雰囲気下に室温で加えた。その混合物を室温で終夜撹拌したのち0℃に冷却して、酢酸エチルで希釈後、5%クエン酸水溶液で中和したのち、セライト濾過をして、濾液を酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(1.98g、22重量%の1,4-ビス(トリメチルシリル)ブタ-1,3-ジインを含む)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ0.26(9H,s),1.50(9H,s),3.31(3H,s),8.55(1H,d,J=1.5Hz),8.99(1H,d,J=1.5Hz)。
Reference Example T004: Preparation of tert-butyl methyl (5-((trimethylsilyl)ethynyl)pyrazin-2-yl)carbamate
Figure JPOXMLDOC01-appb-C000029
tert-butyl (5-iodopyrazin-2-yl)(methyl)carbamate (1.95 g) prepared in Reference Example T003, ethynyltrimethylsilane (1.62 ml) and dichlorobis(triphenylphosphine)palladium(II) (123 mg) ) was added to a mixture of THF (6 ml) and TEA (6 ml) at room temperature under nitrogen atmosphere. The mixture was stirred overnight at room temperature, cooled to 0° C., diluted with ethyl acetate, neutralized with a 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (1.98 g, containing 22% by weight of 1,4-bis(trimethylsilyl)buta-1,3-diyne) as a yellow solid. Ta.
1 H NMR (300 MHz, DMSO-d 6 ) δ 0.26 (9H, s), 1.50 (9H, s), 3.31 (3H, s), 8.55 (1H, d, J=1. 5 Hz), 8.99 (1 H, d, J = 1.5 Hz).
 参考例T005:tert-ブチル (5-エチニルピラジン-2-イル)(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000030
 参考例T004で製造したtert-ブチル メチル(5-((トリメチルシリル)エチニル)ピラジン-2-イル)カルバマート(1.98g、22重量%の1,4-ビス(トリメチルシリル)ブタ-1,3-ジインを含む)のTHF(20ml)の0℃の溶液に、テトラ-n-ブチルアンモニウムフルオリド(1M THF溶液,11.1ml)を加えた。その混合物を0℃で0.5時間撹拌したのち、酢酸エチルと水で希釈して、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(1.11g)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.50(9H,s),3.31(3H,s),4.54(1H,s),8.58(1H,d,J=1.5Hz),8.99(1H,d,J=1.3Hz)。
Reference Example T005: Preparation of tert-butyl (5-ethynylpyrazin-2-yl) (methyl) carbamate
Figure JPOXMLDOC01-appb-C000030
tert-butyl methyl (5-((trimethylsilyl)ethynyl)pyrazin-2-yl)carbamate prepared in Reference Example T004 (1.98 g, 22% by weight of 1,4-bis(trimethylsilyl)buta-1,3-diyne To a 0° C. solution of THF (20 ml) was added tetra-n-butylammonium fluoride (1 M THF solution, 11.1 ml). The mixture was stirred at 0° C. for 0.5 hour, then diluted with ethyl acetate and water and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (1.11 g) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.50 (9H, s), 3.31 (3H, s), 4.54 (1H, s), 8.58 (1H, d, J=1. 5 Hz), 8.99 (1 H, d, J = 1.3 Hz).
 参考例T006:1-(ベンゾ[d]チアゾール-6-イルアミノ)-3-フルオロプロパン-2-オールの製造
Figure JPOXMLDOC01-appb-C000031
 ベンゾ[d]チアゾール-6-アミン(CAS[533-30-2])(1.00g)及び2-(フルオロメチル)オキシラン(CAS[503-09-3])(1.42ml)のMeOH(10ml)溶液を50℃で終夜撹拌後、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(700mg)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ3.01-3.13(1H,m),3.14-3.26(1H,m),3.79-3.99(1H,m),4.27-4.60(2H,m),5.28(1H,d,J=5.3Hz),5.98(1H,t,J=6.0Hz),6.89(1H,dd,J=8.7,2.3Hz),7.16(1H,d,J=2.3Hz),7.75(1H,d,J=8.7Hz),8.90(1H,s)。
Reference Example T006: Preparation of 1-(benzo[d]thiazol-6-ylamino)-3-fluoropropan-2-ol
Figure JPOXMLDOC01-appb-C000031
Benzo[d]thiazol-6-amine (CAS[533-30-2]) (1.00 g) and 2-(fluoromethyl)oxirane (CAS[503-09-3]) (1.42 ml) in MeOH ( 10 ml) The solution was stirred at 50° C. overnight and then concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (700 mg) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 3.01-3.13 (1 H, m), 3.14-3.26 (1 H, m), 3.79-3.99 (1 H, m), 4.27-4.60 (2H, m), 5.28 (1H, d, J = 5.3 Hz), 5.98 (1H, t, J = 6.0 Hz), 6.89 (1H, dd , J = 8.7, 2.3 Hz), 7.16 (1H, d, J = 2.3 Hz), 7.75 (1H, d, J = 8.7 Hz), 8.90 (1H, s) .
 参考例T007:N-(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)ベンゾ[d]チアゾール-6-アミンの製造
Figure JPOXMLDOC01-appb-C000032
 参考例T006で製造した1-(ベンゾ[d]チアゾール-6-イルアミノ)-3-フルオロプロパン-2-オール(700mg)と1H-イミダゾール(316mg)のDMF(15ml)溶液に、tert-ブチルジメチルクロロシラン(606mg)を室温で加えた。その混合物を室温で終夜撹拌し、0℃に冷却後に酢酸エチルと水で希釈して、5%クエン酸水溶液でpH約5としたのち、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(947mg)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ0.03(3H,s),0.03(3H,s),0.86(9H,s),3.04-3.30(2H,m),4.03-4.19(1H,m),4.27-4.62(2H,m),6.00(1H,t,J=6.2Hz),6.86(1H,dd,J=8.8,2.4Hz),7.15(1H,d,J=2.3Hz),7.75(1H,d,J=8.7Hz),8.90(1H,s)。
Reference Example T007: Preparation of N-(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)benzo[d]thiazol-6-amine
Figure JPOXMLDOC01-appb-C000032
In a DMF (15 ml) solution of 1-(benzo[d]thiazol-6-ylamino)-3-fluoropropan-2-ol (700 mg) and 1H-imidazole (316 mg) prepared in Reference Example T006, tert-butyldimethyl Chlorosilane (606 mg) was added at room temperature. The mixture was stirred overnight at room temperature, cooled to 0° C., diluted with ethyl acetate and water, adjusted to pH about 5 with 5% aqueous citric acid solution, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (947 mg) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 0.03 (3H, s), 0.03 (3H, s), 0.86 (9H, s), 3.04-3.30 (2H, m) , 4.03-4.19 (1H, m), 4.27-4.62 (2H, m), 6.00 (1H, t, J = 6.2 Hz), 6.86 (1H, dd, J=8.8, 2.4 Hz), 7.15 (1 H, d, J=2.3 Hz), 7.75 (1 H, d, J=8.7 Hz), 8.90 (1 H, s).
 参考例T008:tert-ブチル ベンゾ[d]チアゾール-6-イル(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000033
 参考例T007で製造したN-(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)ベンゾ[d]チアゾール-6-アミン(945mg)のTHF(15ml)溶液に、二炭酸ジ-tert-ブチル(727mg)とN,N-ジメチルピリジン-4-アミン(170mg)を室温で加えた。その混合物を室温で3時間撹拌後、そこに、二炭酸ジ-tert-ブチル(475mg)を加えた。その混合物を60℃で3時間撹拌後0℃に冷却して、水と酢酸エチルで希釈して、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(1.03g)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ-0.08(3H,s),-0.01(3H,s),0.77(9H,s),1.39(9H,s),3.66-3.88(2H,m),4.05-4.18(1H,m),4.20-4.56(2H,m),7.49(1H,dd,J=8.7,2.3Hz),8.03(1H,d,J=9.0Hz),8.13(1H,d,J=1.9Hz),9.36(1H,s)。
Reference Example T008: Preparation of tert-butyl benzo[d]thiazol-6-yl(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)carbamate
Figure JPOXMLDOC01-appb-C000033
To a solution of N-(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)benzo[d]thiazol-6-amine (945 mg) prepared in Reference Example T007 in THF (15 ml) was added dicarbonic acid. -tert-butyl (727 mg) and N,N-dimethylpyridin-4-amine (170 mg) were added at room temperature. After the mixture was stirred at room temperature for 3 hours, di-tert-butyl dicarbonate (475 mg) was added thereto. The mixture was stirred at 60°C for 3 hours, cooled to 0°C, diluted with water and ethyl acetate and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (1.03 g) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ -0.08 (3H, s), -0.01 (3H, s), 0.77 (9H, s), 1.39 (9H, s), 3.66-3.88 (2H, m), 4.05-4.18 (1H, m), 4.20-4.56 (2H, m), 7.49 (1H, dd, J = 8 .7, 2.3 Hz), 8.03 (1 H, d, J = 9.0 Hz), 8.13 (1 H, d, J = 1.9 Hz), 9.36 (1 H, s).
 参考例T009:tert-ブチル (2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)(2-ホルミルベンゾ[d]チアゾール-6-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000034
 参考例T008で製造したtert-ブチル ベンゾ[d]チアゾール-6-イル(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマート(1.00g)のTHF(18ml)の-78℃の溶液に、n-ブチルリチウム(1.6M ヘキサン溶液、1.70ml)を加えた。その混合物を-78℃で0.5時間撹拌後、そこに、DMF(0.228ml)のTHF(2ml)溶液を加えた。その混合物を-78℃で1.5時間、次いで室温で0.5時間撹拌後、0℃に冷却後に、酢酸エチルで希釈して、5%クエン酸水溶液でpH約5としたのち、水で希釈して、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(166mg)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ-0.09(3H,s),-0.01(3H,s),0.75(9H,s),1.41(9H,s),3.68-3.93(2H,m),4.07-4.22(1H,m),4.22-4.57(2H,m),7.67(1H,dd,J=9.0,2.3Hz),8.23(1H,d,J=8.7Hz),8.26(1H,d,J=1.9Hz),10.10(1H,s)。
Reference Example T009: Preparation of tert-butyl (2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)(2-formylbenzo[d]thiazol-6-yl)carbamate
Figure JPOXMLDOC01-appb-C000034
THF (18 ml) of tert-butyl benzo[d]thiazol-6-yl (2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl) carbamate (1.00 g) prepared in Reference Example T008 - To the solution at 78°C was added n-butyllithium (1.6M hexane solution, 1.70ml). After the mixture was stirred at −78° C. for 0.5 hour, a solution of DMF (0.228 ml) in THF (2 ml) was added thereto. The mixture was stirred at −78° C. for 1.5 hours, then at room temperature for 0.5 hours, cooled to 0° C., diluted with ethyl acetate, adjusted to pH ˜5 with 5% aqueous citric acid, and then diluted with water. Diluted and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (166 mg) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ -0.09 (3H, s), -0.01 (3H, s), 0.75 (9H, s), 1.41 (9H, s), 3.68-3.93 (2H, m), 4.07-4.22 (1H, m), 4.22-4.57 (2H, m), 7.67 (1H, dd, J = 9 .0, 2.3 Hz), 8.23 (1 H, d, J = 8.7 Hz), 8.26 (1 H, d, J = 1.9 Hz), 10.10 (1 H, s).
 参考例T010:tert-ブチル (E)-(2-(2-ブロモビニル)ベンゾ[d]チアゾール-6-イル)(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000035
 (ブロモメチル)トリフェニルホスホニウムブロミド(CAS[1034-49-7])(228mg)のTHF(10ml)の-78℃の溶液に、カリウム tert-ブトキシド(1M THF溶液、0.522ml)を加えた。その混合物を-78℃で2時間撹拌後、そこに、参考例T009で製造したtert-ブチル (2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)(2-ホルミルベンゾ[d]チアゾール-6-イル)カルバマート(163mg)のTHF(5ml)溶液を加えた。その混合物を-78℃から室温で2時間撹拌後、0℃に冷却後に、酢酸エチルと水で希釈して、5%クエン酸水溶液で中和したのち、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(65mg)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ-0.08(3H,s),-0.02(3H,s),0.76(9H,s),1.39(9H,s),3.62-3.87(2H,m),4.05-4.19(1H,m),4.20-4.56(2H,m),7.47(1H,dd,J=8.8,2.1Hz),7.60(1H,d,J=14.1Hz),7.83(1H,d,J=13.9Hz),7.93(1H,d,J=8.7Hz),8.07(1H,d,J=2.3Hz)。
Reference Example T010: of tert-butyl (E)-(2-(2-bromovinyl)benzo[d]thiazol-6-yl)(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)carbamate manufacturing
Figure JPOXMLDOC01-appb-C000035
To a solution of (bromomethyl)triphenylphosphonium bromide (CAS[1034-49-7]) (228 mg) in THF (10 ml) at −78° C. was added potassium tert-butoxide (1 M THF solution, 0.522 ml). After the mixture was stirred at −78° C. for 2 hours, tert-butyl (2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)(2-formylbenzo[d ]thiazol-6-yl)carbamate (163 mg) in THF (5 ml) was added. The mixture was stirred at -78°C to room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate and water, neutralized with 5% aqueous citric acid solution, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (65 mg) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ -0.08 (3H, s), -0.02 (3H, s), 0.76 (9H, s), 1.39 (9H, s), 3.62-3.87 (2H, m), 4.05-4.19 (1H, m), 4.20-4.56 (2H, m), 7.47 (1H, dd, J = 8 .8, 2.1 Hz), 7.60 (1H, d, J=14.1 Hz), 7.83 (1H, d, J=13.9 Hz), 7.93 (1H, d, J=8. 7 Hz), 8.07 (1 H, d, J = 2.3 Hz).
 参考例T011:tert-ブチル (E)-(2-(4-(5-((tert-ブトキシカルボニル)(メチル)アミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000036
 参考例T010で製造したtert-ブチル (E)-(2-(2-ブロモビニル)ベンゾ[d]チアゾール-6-イル)(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマート(30mg)、参考例T005で製造したtert-ブチル (5-エチニルピラジン-2-イル)(メチル)カルバマート(17mg)、及びジクロロビス(トリフェニルホスフィン)パラジウム(II)(1.9mg)のTHF(1ml)とTEA(2ml)との混合液に、ヨウ化銅(I)(1.0mg)を窒素雰囲気下に室温で加えた。その混合物を室温で2時間撹拌したのち、酢酸エチルで希釈後、5%クエン酸水溶液でpH約4としたのち、セライト濾過をして、濾液を酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(22mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ-0.07(3H,s),-0.01(3H,s),0.77(9H,s),1.40(9H,s),1.52(9H,s),3.34(3H,s),3.66-3.88(2H,m),4.06-4.57(3H,m),7.10(1H,d,J=15.8Hz),7.50(1H,brdd,J=8.7,2.3Hz),7.53(1H,d,J=16.2Hz),7.97(1H,d,J=9.0Hz),8.10(1H,d,J=2.3Hz),8.66(1H,d,J=1.5Hz),9.07(1H,d,J=1.5Hz)。
Reference Example T011: tert-butyl (E)-(2-(4-(5-((tert-butoxycarbonyl)(methyl)amino)pyrazin-2-yl)but-1-en-3-yn-1- Preparation of yl)benzo[d]thiazol-6-yl)(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)carbamate
Figure JPOXMLDOC01-appb-C000036
tert-butyl (E)-(2-(2-bromovinyl)benzo[d]thiazol-6-yl)(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl) prepared in Reference Example T010 THF of carbamate (30 mg), tert-butyl (5-ethynylpyrazin-2-yl) (methyl) carbamate (17 mg) prepared in Reference Example T005, and dichlorobis(triphenylphosphine) palladium (II) (1.9 mg) Copper (I) iodide (1.0 mg) was added to a mixture of (1 ml) and TEA (2 ml) at room temperature under a nitrogen atmosphere. The mixture was stirred at room temperature for 2 hours, diluted with ethyl acetate, adjusted to pH about 4 with a 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (22 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ -0.07 (3H, s), -0.01 (3H, s), 0.77 (9H, s), 1.40 (9H, s), 1.52 (9H, s), 3.34 (3H, s), 3.66-3.88 (2H, m), 4.06-4.57 (3H, m), 7.10 (1H, d, J = 15.8 Hz), 7.50 (1H, brdd, J = 8.7, 2.3 Hz), 7.53 (1H, d, J = 16.2 Hz), 7.97 (1H, d , J=9.0 Hz), 8.10 (1H, d, J=2.3 Hz), 8.66 (1H, d, J=1.5 Hz), 9.07 (1H, d, J=1. 5 Hz).
 参考例T012:tert-ブチル (2-ホルミルベンゾ[d]チアゾール-6-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000037
 tert-ブチル ベンゾ[d]チアゾール-6-イルカルバマート(CAS[1192841-91-0])(4.34g)のTHF(50ml)の-78℃の溶液に、n-ブチルリチウム(1.6M ヘキサン溶液、27.1ml)を加えた。その混合物を-78℃で1時間撹拌後、そこに、DMF(5.37ml)のTHF(10ml)溶液を加えた。その混合物を-78℃で1.5時間、次いで室温で0.5時間撹拌後、0℃に冷却にして、酢酸エチルで希釈して、5%クエン酸水溶液でpH約5としたのち、水で希釈して、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣の固体を酢酸エチルに懸濁して、濾取した固体を酢酸エチルで洗浄後に乾燥し、標題化合物(3.24g)を黄色固体として得た。酢酸エチルの濾液と洗浄液を合わせて、シリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(511mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.51(9H,s),7.63(1H,dd,J=9.0,2.2Hz),8.16(1H,d,J=9.3Hz),8.47(1H,d,J=2.0Hz),9.93(1H,s),10.06(1H,s)。
Reference Example T012: Preparation of tert-butyl (2-formylbenzo[d]thiazol-6-yl)carbamate
Figure JPOXMLDOC01-appb-C000037
To a −78° C. solution of tert-butyl benzo[d]thiazol-6-ylcarbamate (CAS[1192841-91-0]) (4.34 g) in THF (50 ml) was added n-butyl lithium (1.6 M hexane solution, 27.1 ml) was added. After the mixture was stirred at -78°C for 1 hour, a solution of DMF (5.37 ml) in THF (10 ml) was added thereto. The mixture was stirred at −78° C. for 1.5 hours, then at room temperature for 0.5 hours, then cooled to 0° C., diluted with ethyl acetate, diluted with 5% aqueous citric acid to pH ˜5, and treated with water. and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residual solid was suspended in ethyl acetate, and the solid collected by filtration was washed with ethyl acetate and dried to give the title compound (3.24 g) as a yellow solid. The ethyl acetate filtrate and washings were combined and purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (511 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.51 (9H, s), 7.63 (1H, dd, J = 9.0, 2.2 Hz), 8.16 (1H, d, J = 9 .3 Hz), 8.47 (1 H, d, J=2.0 Hz), 9.93 (1 H, s), 10.06 (1 H, s).
 参考例T013:tert-ブチル (E)-(2-(2-ブロモビニル)ベンゾ[d]チアゾール-6-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000038
 (ブロモメチル)トリフェニルホスホニウムブロミド(CAS[1034-49-7])(4.23g)のTHF(50ml)の-78℃の溶液に、カリウム tert-ブトキシド(1M THF溶液、8.25ml)を加えた。その混合物を-78℃で2時間撹拌後、そこに、参考例T012で製造したtert-ブチル (2-ホルミルベンゾ[d]チアゾール-6-イル)カルバマート(1.35g)のTHF(15ml)溶液を加えた。その混合物を-78℃で1時間撹拌後、-78℃で酢酸エチルと5%クエン酸水溶液で希釈して、0℃にて5%クエン酸水溶液で中和したのち、水で希釈後に、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(374mg)を淡黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.50(9H,s),7.48(1H,dd,J=9.0,2.2Hz),7.55(1H,d,J=14.0Hz),7.73(1H,d,J=14.0Hz),7.86(1H,d,J=8.9Hz),8.26(1H,d,J=2.0Hz),9.68(1H,s)。
Reference Example T013: Preparation of tert-butyl (E)-(2-(2-bromovinyl)benzo[d]thiazol-6-yl)carbamate
Figure JPOXMLDOC01-appb-C000038
Potassium tert-butoxide (1 M THF solution, 8.25 ml) was added to a solution of (bromomethyl)triphenylphosphonium bromide (CAS[1034-49-7]) (4.23 g) in THF (50 ml) at -78°C. Ta. After the mixture was stirred at -78°C for 2 hours, a solution of tert-butyl (2-formylbenzo[d]thiazol-6-yl)carbamate (1.35g) prepared in Reference Example T012 in THF (15ml) was added thereto. was added. The mixture was stirred at −78° C. for 1 hour, diluted with ethyl acetate and 5% aqueous citric acid solution at −78° C., neutralized with 5% aqueous citric acid solution at 0° C., diluted with water, and then diluted with acetic acid. Extracted with ethyl. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (374 mg) as a pale-yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.50 (9H, s), 7.48 (1H, dd, J = 9.0, 2.2 Hz), 7.55 (1H, d, J = 14 .0Hz), 7.73 (1H, d, J = 14.0Hz), 7.86 (1H, d, J = 8.9Hz), 8.26 (1H, d, J = 2.0Hz), 9 .68 (1H, s).
 参考例T014:tert-ブチル (E)-(5-(4-(6-((tert-ブトキシカルボニル)アミノ)ベンゾ[d]チアゾール-2-イル)ブタ-3-エン-1-イン-1-イル)ピラジン-2-イル(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000039
 参考例T013で製造したtert-ブチル (E)-(2-(2-ブロモビニル)ベンゾ[d]チアゾール-6-イル)カルバマート(370mg)、参考例T005で製造したtert-ブチル (5-エチニルピラジン-2-イル)(メチル)カルバマート(267mg)、及びジクロロビス(トリフェニルホスフィン)パラジウム(II)(37mg)のTHF(6ml)とTEA(3ml)との混合液に、ヨウ化銅(I)(20mg)を窒素雰囲気下に室温で加えた。その混合物を室温で2時間撹拌したのち0℃に冷却して、酢酸エチルで希釈後に、5%クエン酸水溶液でpH約4としたのち、セライト濾過をして、濾液を酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(436mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.50(9H,s),1.51(9H,s),3.34(3H,s),7.01(1H,d,J=16.1Hz),7.48(1H,d,J=16.1Hz),7.50(1H,dd,J=8.9,2.1Hz),7.90(1H,d,J=9.0Hz),8.30(1H,d,J=2.0Hz),8.64(1H,d,J=1.5Hz),9.06(1H,d,J=1.5Hz),9.72(1H,s)。
Reference Example T014: tert-butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yn-1 -yl)pyrazin-2-yl(methyl)carbamate preparation
Figure JPOXMLDOC01-appb-C000039
tert-butyl (E)-(2-(2-bromovinyl)benzo[d]thiazol-6-yl)carbamate (370 mg) prepared in Reference Example T013, tert-butyl prepared in Reference Example T005 (5-ethynylpyrazine -2-yl)(methyl)carbamate (267 mg) and dichlorobis(triphenylphosphine)palladium (II) (37 mg) in THF (6 ml) and TEA (3 ml) was added with copper (I) iodide ( 20 mg) was added at room temperature under a nitrogen atmosphere. The mixture was stirred at room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate, adjusted to pH about 4 with 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (436 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.50 (9H, s), 1.51 (9H, s), 3.34 (3H, s), 7.01 (1H, d, J=16. 1Hz), 7.48 (1H, d, J = 16.1Hz), 7.50 (1H, dd, J = 8.9, 2.1Hz), 7.90 (1H, d, J = 9.0Hz) ), 8.30 (1H, d, J = 2.0 Hz), 8.64 (1H, d, J = 1.5 Hz), 9.06 (1H, d, J = 1.5 Hz), 9.72 (1H, s).
 参考例T015:tert-ブチル (E)-(5-(4-(6-((tert-ブトキシカルボニル)アミノ)ベンゾ[d]チアゾール-2-イル)ブタ-3-エン-1-イン-1-イル)ピリジン-2-イル(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000040
 参考例T013で製造したtert-ブチル (E)-(2-(2-ブロモビニル)ベンゾ[d]チアゾール-6-イル)カルバマート(100mg)、tert-ブチル (5-エチニルピリジン-2-イル)(メチル)カルバマート(CAS[1565797-81-0])(80mg)、及びジクロロビス(トリフェニルホスフィン)パラジウム(II)(9.9mg)のTHF(6ml)とTEA(3ml)との混合液に、ヨウ化銅(I)(5.4mg)を窒素雰囲気下に室温で加えた。その混合物を室温で2時間撹拌したのち0℃に冷却して、酢酸エチルで希釈後に、5%クエン酸水溶液でpH約4としたのち、セライト濾過をして、濾液を酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(140mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.49(9H,s),1.50(9H,s),3.34(3H,s),6.96(1H,d,J=16.1Hz),7.38(1H,d,J=16.0Hz),7.46-7.54(1H,m),7.78(1H,dd,J=8.7,0.8Hz),7.89(1H,brd,J=9.0Hz),7.90(1H,dd,J=8.8,2.3Hz),8.28(1H,d,J=1.3Hz),8.55(1H,dd,J=2.4,0.8Hz),9.71(1H,s)。
Reference Example T015: tert-butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yn-1 -yl)pyridin-2-yl(methyl)carbamate preparation
Figure JPOXMLDOC01-appb-C000040
tert-butyl (E)-(2-(2-bromovinyl)benzo[d]thiazol-6-yl)carbamate (100 mg) prepared in Reference Example T013, tert-butyl (5-ethynylpyridin-2-yl) ( methyl)carbamate (CAS[1565797-81-0]) (80 mg) and dichlorobis(triphenylphosphine)palladium (II) (9.9 mg) in THF (6 ml) and TEA (3 ml) was added with iodine. Cupric (I) (5.4 mg) was added at room temperature under a nitrogen atmosphere. The mixture was stirred at room temperature for 2 hours, cooled to 0°C, diluted with ethyl acetate, adjusted to pH about 4 with 5% aqueous citric acid solution, filtered through celite, and the filtrate was extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (140 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.49 (9H, s), 1.50 (9H, s), 3.34 (3H, s), 6.96 (1H, d, J=16. 1Hz), 7.38 (1H, d, J = 16.0Hz), 7.46-7.54 (1H, m), 7.78 (1H, dd, J = 8.7, 0.8Hz), 7.89 (1H, brd, J = 9.0Hz), 7.90 (1H, dd, J = 8.8, 2.3Hz), 8.28 (1H, d, J = 1.3Hz), 8 .55 (1 H, dd, J=2.4, 0.8 Hz), 9.71 (1 H, s).
 参考例T016:(E)-2-(4-(6-(メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-アミンの製造
Figure JPOXMLDOC01-appb-C000041
 参考例T015で製造したtert-ブチル (E)-(5-(4-(6-((tert-ブトキシカルボニル)アミノ)ベンゾ[d]チアゾール-2-イル)ブタ-3-エン-1-イン-1-イル)ピリジン-2-イル(メチル)カルバマート(140mg)、TFA(0.900ml)及び水(0.100ml)の混合物を室温で1時間撹拌後、0℃に冷却して、酢酸エチルで希釈後に5%炭酸水素ナトリウム水溶液で中和した後、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮し、標題化合物(84mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.80(3H,d,J=4.9Hz),5.58(2H,s),6.47(1H,d,J=8.3Hz),6.71(1H,d,J=15.8Hz),6.77(1H,dd,J=8.7,2.3Hz),7.03(1H,d,J=2.3Hz),7.05-7.11(1H,m),7.08-7.16(1H,m),7.43-7.50(1H,m),7.61(1H,d,J=8.7Hz),8.17(1H,d,J=2.3Hz)。
Reference Example T016: (E)-2-(4-(6-(methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-amine manufacturing
Figure JPOXMLDOC01-appb-C000041
tert-butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yne prepared in Reference Example T015 A mixture of 1-yl)pyridin-2-yl(methyl)carbamate (140 mg), TFA (0.900 ml) and water (0.100 ml) was stirred at room temperature for 1 hour, then cooled to 0°C and ethyl acetate and neutralized with 5% aqueous sodium bicarbonate, extracted with ethyl acetate, washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The title compound (84 mg) was obtained as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.80 (3H, d, J = 4.9 Hz), 5.58 (2H, s), 6.47 (1H, d, J = 8.3 Hz), 6.71 (1H, d, J = 15.8Hz), 6.77 (1H, dd, J = 8.7, 2.3Hz), 7.03 (1H, d, J = 2.3Hz), 7 .05-7.11 (1H, m), 7.08-7.16 (1H, m), 7.43-7.50 (1H, m), 7.61 (1H, d, J=8. 7 Hz), 8.17 (1 H, d, J = 2.3 Hz).
 参考例T017:2-ヒドロキシ-3-ヨードプロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000042
 3-ヨードプロパン-1,2-ジオール(CAS[554-10-9])(3.40g)のピリジン(12ml)溶液に、塩化パラトルエンスルホニル(3.21g)を室温で加えた。その混合物を室温で4時間撹拌後、0℃に冷却して、酢酸エチルと1M塩酸で希釈後、酢酸エチルで抽出した。抽出液を1M塩酸、水及び食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製して標題化合物(3.95g)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ2.43(3H,s),3.14(1H,dd,J=10.3,5.6Hz),3.21(1H,dd,J=10.3,5.3Hz),3.57-3.69(1H,m),3.91(1H,dd,J=9.9,6.0Hz),3.97(1H,dd,J=9.9,4.4Hz),5.71(1H,d,J=5.3Hz),7.50(2H,dd,J=8.6,0.7Hz),7.80(2H,d,J=8.3Hz)。
Reference Example T017: Preparation of 2-hydroxy-3-iodopropyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000042
To a solution of 3-iodopropane-1,2-diol (CAS[554-10-9]) (3.40 g) in pyridine (12 ml) was added p-toluenesulfonyl chloride (3.21 g) at room temperature. The mixture was stirred at room temperature for 4 hours, cooled to 0° C., diluted with ethyl acetate and 1M hydrochloric acid, and extracted with ethyl acetate. The extract was washed with 1M hydrochloric acid, water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (3.95 g) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.43 (3H, s), 3.14 (1H, dd, J = 10.3, 5.6 Hz), 3.21 (1H, dd, J = 10 .3, 5.3 Hz), 3.57-3.69 (1H, m), 3.91 (1H, dd, J = 9.9, 6.0 Hz), 3.97 (1H, dd, J = 9.9, 4.4 Hz), 5.71 (1H, d, J = 5.3 Hz), 7.50 (2H, dd, J = 8.6, 0.7 Hz), 7.80 (2H, d , J=8.3 Hz).
 参考例T018:3-ヨード-2-((テトラヒドロ-2H-ピラン-2-イル)オキシ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000043
 参考例T017で製造した(2-ヒドロキシ-3-ヨードプロピル 4-メチルベンゼンスルホナート(2.00g)及び3,4-ジヒドロ-2H-ピラン(5.12ml)のTHF(25ml)溶液に、パラトルエンスルホン酸ピリジニウム(1.41g)を室温で加えた。その混合物を室温で4時間撹拌したのち、パラトルエンスルホン酸一水和物(534mg)を室温で加えた。その混合物を室温で4時間撹拌したのち、0℃に冷却して、酢酸エチルで希釈後、5%炭酸水素ナトリウム水溶液で中和して、水で希釈後に酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(2.09g)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ1.28-1.73(6H,m),2.43(3H,s),3.27-3.47(3H,m),3.56-3.89(2H,m),4.04-4.20(2H,m),4.59-4.78(1H,m),7.50(2H,d,J=8.0Hz),7.81(2H,dd,J=8.3,1.3Hz)。
Reference Example T018: Preparation of 3-iodo-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000043
To a THF (25 ml) solution of (2-hydroxy-3-iodopropyl 4-methylbenzenesulfonate (2.00 g) and 3,4-dihydro-2H-pyran (5.12 ml) prepared in Reference Example T017, para Pyridinium toluenesulfonate (1.41 g) was added at room temperature, the mixture was stirred at room temperature for 4 hours, para-toluenesulfonic acid monohydrate (534 mg) was added at room temperature, and the mixture was stirred at room temperature for 4 hours. After stirring, the mixture was cooled to 0° C., diluted with ethyl acetate, neutralized with a 5% aqueous sodium hydrogencarbonate solution, diluted with water and extracted with ethyl acetate.The extract was washed with water and brine, The extract was dried over anhydrous sodium sulfate, concentrated under reduced pressure, and purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (2.09 g) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.28-1.73 (6H, m), 2.43 (3H, s), 3.27-3.47 (3H, m), 3.56- 3.89 (2H, m), 4.04-4.20 (2H, m), 4.59-4.78 (1H, m), 7.50 (2H, d, J=8.0Hz), 7.81 (2H, dd, J=8.3, 1.3 Hz).
 参考例T019:3-ヒドロキシ-2,2-ジメトキシプロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000044
 2,2-ジメトキシプロパン-1,3-ジオール(CAS[153214-82-5])(2.28g)のピリジン(6ml)溶液に、塩化パラトルエンスルホニル(1.00g)をゆっくり0℃で加えた。その混合物を0℃から室温で3日間撹拌後、0℃に冷却して、酢酸エチル、水及び1M塩酸で希釈後、酢酸エチルで抽出した。抽出液を水及び食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製して標題化合物(389mg)を白色固体として得た。
H NMR(300MHz,DMSO-d)δ2.43(3H,s),3.04(6H,s),3.32(2H,brd,J=5.5Hz),3.89(2H,s),4.92(1H,t,J=5.6Hz),7.49(2H,d,J=8.0Hz),7.82(2H,d,J=8.3Hz)。
Reference Example T019: Preparation of 3-hydroxy-2,2-dimethoxypropyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000044
To a pyridine (6 ml) solution of 2,2-dimethoxypropane-1,3-diol (CAS [153214-82-5]) (2.28 g) was slowly added p-toluenesulfonyl chloride (1.00 g) at 0°C. Ta. The mixture was stirred at 0°C to room temperature for 3 days, cooled to 0°C, diluted with ethyl acetate, water and 1M hydrochloric acid, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (389 mg) as a white solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.43 (3H, s), 3.04 (6H, s), 3.32 (2H, brd, J = 5.5 Hz), 3.89 (2H, s), 4.92 (1H, t, J = 5.6 Hz), 7.49 (2H, d, J = 8.0 Hz), 7.82 (2H, d, J = 8.3 Hz).
 参考例T020:2,2-ジメトキシ-3-オキソプロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000045
 参考例T019で製造した3-ヒドロキシ-2,2-ジメトキシプロピル 4-メチルベンゼンスルホナート(389mg)のアセトニトリル(20ml)溶液に、1,1,1-トリアセトキシ-1,1-ジヒドロ-1,2-ベンゾヨードキソール-3-(1H)-オン(1.14g)を室温で加えた。その混合物を室温で2時間撹拌したのち、0℃に冷却し、酢酸エチルと飽和チオ硫酸ナトリウム水溶液で希釈し、5%炭酸水素ナトリウム水溶液で中和した後、酢酸エチルで抽出した。抽出液を5%炭酸水素ナトリウム水溶液、水、食塩水の順で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(336mg)を無色液体として得た。
H NMR(300MHz,DMSO-d)δ2.43(3H,s),3.18(6H,s),4.14(2H,s),7.50(2H,dd,J=8.6,0.7Hz),7.79(2H,d,J=8.3Hz),9.44(1H,s)。
Reference Example T020: Preparation of 2,2-dimethoxy-3-oxopropyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000045
1,1,1-Triacetoxy-1,1-dihydro-1,1,1-triacetoxy-1,1-dihydro-1,1,1-triacetoxy-1,1-dihydro-1, 2-benzoiodoxol-3-(1H)-one (1.14 g) was added at room temperature. The mixture was stirred at room temperature for 2 hours, then cooled to 0°C, diluted with ethyl acetate and saturated aqueous sodium thiosulfate, neutralized with 5% aqueous sodium hydrogen carbonate and extracted with ethyl acetate. The extract was washed with a 5% aqueous sodium hydrogen carbonate solution, water and brine in that order, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (336 mg) as a colorless liquid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.43 (3H, s), 3.18 (6H, s), 4.14 (2H, s), 7.50 (2H, dd, J=8. 6, 0.7 Hz), 7.79 (2H, d, J = 8.3 Hz), 9.44 (1 H, s).
 参考例T021:(E)-2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-アミンの製造
Figure JPOXMLDOC01-appb-C000046
 参考例T014で製造したtert-ブチル (E)-(5-(4-(6-((tert-ブトキシカルボニル)アミノ)ベンゾ[d]チアゾール-2-イル)ブタ-3-エン-1-イン-1-イル)ピラジン-2-イル(メチル)カルバマート(736mg)、TFA(2.70ml)及び水(0.300ml)の混合物を室温で1時間撹拌後、0℃に冷却して、酢酸エチルで希釈後に5%炭酸水素ナトリウム水溶液で中和した後、酢酸エチルとTHFの混合溶媒で抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮し、標題化合物(446mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.84(3H,d,J=4.9Hz),5.60(2H,s),6.74(1H,d,J=16.1Hz),6.74-6.83(1H,m),7.04(1H,d,J=2.2Hz),7.20(1H,d,J=16.0Hz),7.57-7.64(1H,m),7.63(1H,d,J=8.8Hz),7.93(1H,d,J=1.4Hz),8.19(1H,d,J=1.3Hz)。
Reference Example T021: (E)-2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-amine manufacturing
Figure JPOXMLDOC01-appb-C000046
tert-Butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yne prepared in Reference Example T014 A mixture of 1-yl)pyrazin-2-yl(methyl)carbamate (736 mg), TFA (2.70 ml) and water (0.300 ml) was stirred at room temperature for 1 hour, then cooled to 0° C. and ethyl acetate and neutralized with a 5% aqueous sodium hydrogencarbonate solution, extracted with a mixed solvent of ethyl acetate and THF, washed with water and brine, dried over anhydrous sodium sulfate, and concentrated under reduced pressure. , to give the title compound (446 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.84 (3H, d, J = 4.9 Hz), 5.60 (2H, s), 6.74 (1H, d, J = 16.1 Hz), 6.74-6.83 (1H, m), 7.04 (1H, d, J = 2.2Hz), 7.20 (1H, d, J = 16.0Hz), 7.57-7.64 (1H, m), 7.63 (1H, d, J = 8.8Hz), 7.93 (1H, d, J = 1.4Hz), 8.19 (1H, d, J = 1.3Hz) .
 参考例T022:(E)-2,2-ジメトキシ-3-((2-((E)-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)イミノ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000047
 参考例T020で製造した2,2-ジメトキシ-3-オキソプロピル 4-メチルベンゼンスルホナート(560mg)及び参考例T021で製造した(E)-2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-アミン(552mg)の酢酸エチル(60ml)及びTHF(20ml)の懸濁液を、室温で2時間撹拌後、さらにTHF(80ml)を加え、この溶液を40℃で2時間、室温で終夜撹拌した。撹拌後、減圧下で濃縮し、標題化合物(1.05g)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.30(3H,s),2.85(3H,d,J=4.9Hz),3.24(6H,s),4.29(2H,s),7.07(1H,d,J=16.0Hz),7.20(1H,dd,J=8.7,2.2Hz),7.36(1H,d,J=16.0Hz),7.36(2H,d,J=7.9Hz),7.69(1H,q,J=4.9Hz),7.75-7.79(3H,m),7.87(1H,s),7.95(1H,d,J=1.5Hz),7.97(1H,d,J=8.9Hz),8.23(1H,d,J=1.4Hz)。
Reference Example T022: (E)-2,2-dimethoxy-3-((2-((E)-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yne) Preparation of 1-yl)benzo[d]thiazol-6-yl)imino)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000047
2,2-dimethoxy-3-oxopropyl 4-methylbenzenesulfonate (560 mg) prepared in Reference Example T020 and (E)-2-(4-(5-(methylamino)pyrazine- A suspension of 2-yl)but-1-en-3-yn-1-yl)benzo[d]thiazol-6-amine (552 mg) in ethyl acetate (60 ml) and THF (20 ml) was treated at room temperature for 2 After stirring for an hour, more THF (80 ml) was added and the solution was stirred at 40° C. for 2 hours and at room temperature overnight. After stirring, the mixture was concentrated under reduced pressure to give the title compound (1.05 g) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.30 (3H, s), 2.85 (3H, d, J = 4.9 Hz), 3.24 (6H, s), 4.29 (2H, s), 7.07 (1H, d, J = 16.0 Hz), 7.20 (1H, dd, J = 8.7, 2.2 Hz), 7.36 (1H, d, J = 16.0 Hz ), 7.36 (2H, d, J = 7.9Hz), 7.69 (1H, q, J = 4.9Hz), 7.75-7.79 (3H, m), 7.87 (1H , s), 7.95 (1 H, d, J = 1.5 Hz), 7.97 (1 H, d, J = 8.9 Hz), 8.23 (1 H, d, J = 1.4 Hz).
 参考例T023:(E)-2,2-ジメトキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000048
 参考例T022で製造した(E)-2,2-ジメトキシ-3-((2-((E)-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)イミノ)プロピル 4-メチルベンゼンスルホナート(1.05g)のTHF(40ml)の懸濁液に水素化ホウ素ナトリウム(69mg)のDMSO(10ml)の溶液を加え、次にEtOH(20ml)を0℃で加えた。その混合物を0℃で1時間撹拌後、酢酸エチルと水で希釈して、5%クエン酸水溶液でpH約4の酸性にした後、酢酸エチルで抽出した。抽出液を水及び食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(935mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.20(3H,s),2.84(3H,d,J=4.8Hz),3.12-3.16(2H,m),3.14(6H,s),4.02(2H,s),5.73(1H,t,J=5.1Hz),6.80(1H,d,J=16.1Hz),6.85(1H,dd,J=9.0,2.2Hz),7.01(1H,d,J=2.3Hz),7.16-7.27(3H,m),7.61-7.70(4H,m),7.94(1H,d,J=1.5Hz),8.20(1H,d,J=1.4Hz)。
Reference Example T023: (E)-2,2-dimethoxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) Preparation of benzo[d]thiazol-6-yl)amino)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000048
(E)-2,2-dimethoxy-3-((2-((E)-(4-(5-(methylamino)pyrazin-2-yl)but-1-ene-3) prepared in Reference Example T022 -yn-1-yl)benzo[d]thiazol-6-yl)imino)propyl 4-methylbenzenesulfonate (1.05g) in THF (40ml) in suspension in sodium borohydride (69mg) in DMSO (10 ml) was added followed by EtOH (20 ml) at 0° C. The mixture was stirred at 0° C. for 1 hour and then diluted with ethyl acetate and water and diluted with 5% aqueous citric acid to pH ˜4. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure, and the residue was subjected to silica gel column chromatography (hexane/ethyl acetate). Purification gave the title compound (935 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.20 (3H, s), 2.84 (3H, d, J = 4.8 Hz), 3.12-3.16 (2H, m), 3. 14 (6H, s), 4.02 (2H, s), 5.73 (1H, t, J = 5.1 Hz), 6.80 (1H, d, J = 16.1 Hz), 6.85 ( 1H, dd, J = 9.0, 2.2 Hz), 7.01 (1H, d, J = 2.3 Hz), 7.16-7.27 (3H, m), 7.61-7.70 (4H,m), 7.94 (1H,d,J=1.5Hz), 8.20 (1H,d,J=1.4Hz).
 参考例T024:(E)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)-2-オキソプロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000049
 参考例T023で製造した(E)-2,2-ジメトキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロピル 4-メチルベンゼンスルホナート(132mg)、TFA(1.35ml)及び水(0.15ml)の混合物を室温で1時間撹拌後、0℃に冷却して、酢酸エチルで希釈後に5%炭酸水素ナトリウム水溶液で中和した後、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して、減圧下で濃縮した。残渣の固体を酢酸エチルに懸濁してろ取した後、酢酸エチルで洗浄後に乾燥して、標題化合物(83mg)を橙色固体として得た。
H NMR(300MHz,DMSO-d)δ2.41(3H,s),2.84(3H,d,J=4.9Hz),4.11(2H,d,J=5.9Hz),5.01(2H,s),6.45(1H,t,J=5.7Hz),6.79(1H,d,J=16.0Hz),6.85(1H,dd,J=9.0,2.4Hz),6.96(1H,d,J=2.3Hz),7.22(1H,d,J=16.0Hz),7.47(2H,d,J=7.9Hz),7.62(1H,q,J=6.0Hz),7.67(1H,d,J=8.9Hz),7.80(2H,d,J=8.4Hz),7.93(1H,d,J=1.4Hz),8.19(1H,d,J=1.3Hz)。
Reference Example T024: (E)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d]thiazole- Preparation of 6-yl)amino)-2-oxopropyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000049
(E)-2,2-dimethoxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-) prepared in Reference Example T023 A mixture of yl)benzo[d]thiazol-6-yl)amino)propyl 4-methylbenzenesulfonate (132 mg), TFA (1.35 ml) and water (0.15 ml) was stirred at room temperature for 1 hour, then stirred at 0°C. The mixture was cooled to , diluted with ethyl acetate, neutralized with a 5% aqueous sodium hydrogencarbonate solution, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residual solid was suspended in ethyl acetate and collected by filtration, washed with ethyl acetate and dried to give the title compound (83 mg) as an orange solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.41 (3H, s), 2.84 (3H, d, J = 4.9 Hz), 4.11 (2H, d, J = 5.9 Hz), 5.01 (2H, s), 6.45 (1H, t, J = 5.7 Hz), 6.79 (1H, d, J = 16.0 Hz), 6.85 (1H, dd, J = 9 .0, 2.4 Hz), 6.96 (1H, d, J = 2.3 Hz), 7.22 (1H, d, J = 16.0 Hz), 7.47 (2H, d, J = 7.0 Hz). 9 Hz), 7.62 (1 H, q, J = 6.0 Hz), 7.67 (1 H, d, J = 8.9 Hz), 7.80 (2 H, d, J = 8.4 Hz), 7. 93 (1 H, d, J = 1.4 Hz), 8.19 (1 H, d, J = 1.3 Hz).
 参考例T025:(E)-2-ヒドロキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000050
 参考例T024で製造した(E)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)-2-オキソプロピル 4-メチルベンゼンスルホナート(83mg)のEtOH(20ml)及びTHF(20ml)の懸濁液に水素化ホウ素ナトリウム(8.8mg)を室温で加えた。その懸濁液を超音波処理で溶液にした後、室温で10時間撹拌した。撹拌後、0℃に冷却して酢酸エチルと水で希釈後に、5%クエン酸水溶液でpH約4の酸性にした後、酢酸エチルで抽出した。抽出液を水及び食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(50mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.36(3H,s),2.84(3H,d,J=4.9Hz),3.00-3.17(2H,m),3.76-3.89(1H,m),3.96(1H,dd,J=10.1,6.3Hz),4.08(1H,dd,J=9.7,3.7Hz),5.37(1H,d,J=5.1Hz),6.13(1H,t,J=6.4Hz),6.78(1H,d,J=16.1Hz),6.81(1H,dd,J=8.6,2.6Hz),7.01(1H,d,J=2.1Hz),7.21(1H,d,J=16.0Hz),7.40(2H,d,J=7.9Hz),7.62(1H,q,J=5.5Hz),7.65(1H,d,J=9.0Hz),7.76(2H,d,J=8.3Hz),7.93(1H,d,J=1.4Hz),8.19(1H,d,J=1.3Hz)。
Reference Example T025: (E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[ Preparation of d]thiazol-6-yl)amino)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000050
(E)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d] prepared in Reference Example T024 Sodium borohydride (8.8 mg) was added to a suspension of thiazol-6-yl)amino)-2-oxopropyl 4-methylbenzenesulfonate (83 mg) in EtOH (20 ml) and THF (20 ml) at room temperature. Ta. The suspension was sonicated into a solution and then stirred at room temperature for 10 hours. After stirring, the mixture was cooled to 0° C., diluted with ethyl acetate and water, acidified to about pH 4 with a 5% aqueous citric acid solution, and then extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (50 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.36 (3H, s), 2.84 (3H, d, J = 4.9 Hz), 3.00-3.17 (2H, m), 3. 76-3.89 (1H, m), 3.96 (1H, dd, J = 10.1, 6.3Hz), 4.08 (1H, dd, J = 9.7, 3.7Hz), 5 .37 (1H, d, J = 5.1 Hz), 6.13 (1H, t, J = 6.4 Hz), 6.78 (1H, d, J = 16.1 Hz), 6.81 (1H, dd, J = 8.6, 2.6 Hz), 7.01 (1H, d, J = 2.1 Hz), 7.21 (1H, d, J = 16.0 Hz), 7.40 (2H, d , J=7.9 Hz), 7.62 (1H, q, J=5.5 Hz), 7.65 (1 H, d, J=9.0 Hz), 7.76 (2H, d, J=8. 3 Hz), 7.93 (1 H, d, J = 1.4 Hz), 8.19 (1 H, d, J = 1.3 Hz).
 実施例1:(E)-1-フルオロ-3-((2-(4-(5-メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オール(化合物(I)。以下、「SPAL-T-83」ともいう)の製造
Figure JPOXMLDOC01-appb-C000051
 参考例T011で製造したtert-ブチル (E)-(2-(4-(5-((tert-ブトキシカルボニル)(メチル)アミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)(2-((tert-ブチルジメチルシリル)オキシ)-3-フルオロプロピル)カルバマート(22mg)、TFA(0.900ml)及び水(0.100ml)の混合物を室温で4時間撹拌後、0℃に冷却して、酢酸エチルで希釈後に5%炭酸水素ナトリウム水溶液で中和した後、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、短いシリカゲルカラムを通して減圧下で濃縮した。残渣の固体を酢酸エチルに懸濁させ、濾取した固体を酢酸エチルで洗浄後に乾燥し、標題化合物(11.5mg)を橙色固体として得た。
H NMR(300MHz,DMSO-d)δ2.84(3H,d,J=4.9Hz),3.02-3.15(1H,m),3.16-3.28(1H,m),3.81-3.98(1H,m),4.28-4.58(2H,m),5.29(1H,d,J=5.3Hz),6.20(1H,t,J=6.0Hz),6.76(1H,d,J=15.8Hz),6.88(1H,dd,J=9.0,2.3Hz),7.10(1H,d,J=2.3Hz),7.21(1H,d,J=16.2Hz),7.61(1H,q,J=4.9Hz),7.67(1H,d,J=9.0Hz),7.93(1H,d,J=1.5Hz),8.19(1H,d,J=1.5Hz)。
Example 1: (E)-1-fluoro-3-((2-(4-(5-methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d ] Production of thiazol-6-yl)amino)propan-2-ol (Compound (I), hereinafter also referred to as “SPAL-T-83”)
Figure JPOXMLDOC01-appb-C000051
tert-Butyl (E)-(2-(4-(5-((tert-butoxycarbonyl)(methyl)amino)pyrazin-2-yl)but-1-en-3-yn- prepared in Reference Example T011 1-yl)benzo[d]thiazol-6-yl)(2-((tert-butyldimethylsilyl)oxy)-3-fluoropropyl)carbamate (22 mg), TFA (0.900 ml) and water (0.100 ml) ) was stirred at room temperature for 4 hours, cooled to 0° C., diluted with ethyl acetate, neutralized with 5% aqueous sodium hydrogen carbonate solution, and extracted with ethyl acetate. The extract was washed with water and brine, dried over anhydrous sodium sulfate, passed through a short silica gel column and concentrated under reduced pressure. The residual solid was suspended in ethyl acetate, and the solid collected by filtration was washed with ethyl acetate and dried to give the title compound (11.5 mg) as an orange solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.84 (3H, d, J = 4.9 Hz), 3.02-3.15 (1H, m), 3.16-3.28 (1H, m ), 3.81-3.98 (1H, m), 4.28-4.58 (2H, m), 5.29 (1H, d, J = 5.3 Hz), 6.20 (1H, t , J = 6.0 Hz), 6.76 (1H, d, J = 15.8 Hz), 6.88 (1H, dd, J = 9.0, 2.3 Hz), 7.10 (1H, d, J = 2.3Hz), 7.21 (1H, d, J = 16.2Hz), 7.61 (1H, q, J = 4.9Hz), 7.67 (1H, d, J = 9.0Hz) ), 7.93 (1 H, d, J=1.5 Hz), 8.19 (1 H, d, J=1.5 Hz).
 実施例2:(E)-1-フルオロ-3-((2-(4-(6-メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロパン-2-オール(化合物(II)。以下、「SPAL-T-95」ともいう)の製造
Figure JPOXMLDOC01-appb-C000052
 参考例T016で製造した(E)-2-(4-(6-(メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-アミン(40mg)、2-(フルオロメチル)オキシラン(CAS[503-09-3])(0.047ml)及びメトキシエタン-1-オール(6ml)の混合物をマイクロウェーブ照射下に150℃で1時間撹拌した。反応混合物を室温に冷却後、そこに、2-(フルオロメチル)オキシラン(0.047ml)を加えてマイクロウェーブ照射下で150℃で1時間撹拌し、室温に冷却後に減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(7.0mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.80(3H,d,J=4.9Hz),3.01-3.28(2H,m),3.81-3.97(1H,m),4.28-4.58(2H,m),5.30(1H,d,J=5.0Hz),6.18(1H,t,J=6.1Hz),6.47(1H,d,J=8.7Hz),6.73(1H,d,J=16.0Hz),6.87(1H,dd,J=8.8,2.3Hz),7.03-7.14(2H,m),7.12(1H,d,J=16.0Hz),7.47(1H,dd,J=8.8,2.4Hz),7.65(1H,d,J=8.9Hz),8.17(1H,d,J=2.3Hz)。
Example 2: (E)-1-fluoro-3-((2-(4-(6-methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d ] Production of thiazol-6-yl)amino)propan-2-ol (compound (II), hereinafter also referred to as “SPAL-T-95”)
Figure JPOXMLDOC01-appb-C000052
(E)-2-(4-(6-(methylamino)pyridin-3-yl)but-1-en-3-yn-1-yl)benzo[d]thiazole-6- prepared in Reference Example T016 A mixture of amine (40 mg), 2-(fluoromethyl)oxirane (CAS [503-09-3]) (0.047 ml) and methoxyethan-1-ol (6 ml) was heated at 150° C. for 1 hour under microwave irradiation. Stirred. After cooling the reaction mixture to room temperature, 2-(fluoromethyl)oxirane (0.047 ml) was added thereto and stirred at 150° C. for 1 hour under microwave irradiation. After cooling to room temperature, the mixture was concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (7.0 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.80 (3H, d, J = 4.9 Hz), 3.01-3.28 (2H, m), 3.81-3.97 (1H, m ), 4.28-4.58 (2H, m), 5.30 (1H, d, J = 5.0 Hz), 6.18 (1H, t, J = 6.1 Hz), 6.47 (1H , d, J=8.7 Hz), 6.73 (1H, d, J=16.0 Hz), 6.87 (1H, dd, J=8.8, 2.3 Hz), 7.03-7. 14 (2H, m), 7.12 (1H, d, J = 16.0Hz), 7.47 (1H, dd, J = 8.8, 2.4Hz), 7.65 (1H, d, J = 8.9 Hz), 8.17 (1H, d, J = 2.3 Hz).
 実施例3:(E)-3((tert-ブトキシカルボニル)(2-(4-(5-((tert-ブトキシカルボニル)(メチル)アミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)-2-((テトラヒドロ-2H-ピラン-2-イル)オキシ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000053
 参考例T014で製造したtert-ブチル (E)-(5-(4-(6-((tert-ブトキシカルボニル)アミノ)ベンゾ[d]チアゾール-2-イル)ブタ-3-エン-1-イン-1-イル)ピラジン-2-イル(メチル)カルバマート(436mg)のDMF(20ml)の0℃の溶液に、水素化ナトリウム(60%油性、41mg)を加えた。その混合物を0℃で5分撹拌後、そこに、参考例T018で製造した3-ヨード-2-((テトラヒドロ-2H-ピラン-2-イル)オキシ)プロピル 4-メチルベンゼンスルホナート(567mg)のDMF(5ml)溶液を加えた。その混合物を室温で2時間撹拌後、0℃に冷却して、酢酸エチルと水で希釈後に5%クエン酸水溶液で中和して、酢酸エチルで抽出した。抽出液を水と食塩水で洗浄後、無水硫酸ナトリウムで乾燥して、減圧下で濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(42mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ1.23-1.43(6H,m),1.37(9H,s),1.52(9H,s),2.41(3H,s),3.18-3.28(1H,m),3.34(3H,s),3.51-3.64(1H,m),3.69-3.85(2H,m),3.85-3.96(1H,m),3.98-4.10(2H,m),4.50(1H,brs),7.12(1H,d,J=16.1Hz),7.30-7.47(1H,m),7.43(2H,brd,J=8.1Hz),7.54(1H,d,J=16.1Hz),7.66-7.79(2H,m),7.94(1H,d,J=8.7Hz),7.98-8.05(1H,m),8.66(1H,d,J=1.4Hz),9.07(1H,d,J=1.3Hz)。
Example 3: (E)-3((tert-butoxycarbonyl)(2-(4-(5-((tert-butoxycarbonyl)(methyl)amino)pyrazin-2-yl)but-1-ene-3 -yn-1-yl)benzo[d]thiazol-6-yl)amino)-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate Preparation
Figure JPOXMLDOC01-appb-C000053
tert-Butyl (E)-(5-(4-(6-((tert-butoxycarbonyl)amino)benzo[d]thiazol-2-yl)but-3-en-1-yne prepared in Reference Example T014 -1-yl)pyrazin-2-yl(methyl)carbamate (436 mg) in DMF (20 ml) at 0° C. was added sodium hydride (60% oily, 41 mg). After stirring for a minute, a DMF (5 ml) solution of 3-iodo-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate (567 mg) prepared in Reference Example T018 was added thereto. The mixture was stirred at room temperature for 2 hours, cooled to 0° C., diluted with ethyl acetate and water, neutralized with 5% aqueous citric acid solution, and extracted with ethyl acetate. After washing with water, it was dried over anhydrous sodium sulfate, concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (42 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 1.23-1.43 (6H, m), 1.37 (9H, s), 1.52 (9H, s), 2.41 (3H, s) , 3.18-3.28 (1H, m), 3.34 (3H, s), 3.51-3.64 (1H, m), 3.69-3.85 (2H, m), 3 .85-3.96 (1H, m), 3.98-4.10 (2H, m), 4.50 (1H, brs), 7.12 (1H, d, J = 16.1 Hz), 7 .30-7.47 (1H, m), 7.43 (2H, brd, J = 8.1 Hz), 7.54 (1H, d, J = 16.1 Hz), 7.66-7.79 ( 2H, m), 7.94 (1H, d, J=8.7 Hz), 7.98-8.05 (1H, m), 8.66 (1H, d, J=1.4 Hz), 9. 07 (1H, d, J = 1.3 Hz).
 実施例4:(E)-3-(2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)オキサゾリジン-5-イル)メチル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000054
 参考例T025で製造した(E)-2-ヒドロキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)ベンゾ[d]チアゾール-6-イル)アミノ)プロピル 4-メチルベンゼンスルホナート(375mg)とパラホルムアルデヒド(63mg)のアセトニトリル(80ml)混合物を60℃で0.5時間撹拌後、パラホルムアルデヒド(42mg)を加えて、さらに60℃で0.5時間撹拌して室温まで冷却し、短いシリカゲルカラムを通して酢酸エチルで溶出し、減圧下に濃縮した。残渣をシリカゲルカラムクロマトグラフィー(ヘキサン/酢酸エチル)で精製し、標題化合物(166mg)を黄色固体として得た。
H NMR(300MHz,DMSO-d)δ2.42(3H,s),2.84(3H,d,J=4.8Hz),3.19(1H,dd,J=9.2,6.5Hz),3.55(1H,dd,J=9.4,7.3Hz),4.19(1H,dd,J=11.0,6.1Hz),4.24(1H,dd,J=10.8,3.5Hz),4.49-4.61(1H,m),4.84(1H,d,J=2.8Hz),4.89(1H,d,J=2.8Hz),6.77(1H,dd,J=9.1,2.4Hz),6.85(1H,d,J=16.1Hz),7.13(1H,d,J=2.4Hz),7.25(1H,d,J=16.1Hz),7.48(2H,d,J=7.9Hz),7.64(1H,q,J=4.6Hz),7.77-7.85(3H,m),7.94(1H,d,J=1.5Hz),8.20(1H,d,J=1.4Hz)。
Example 4: (E)-3-(2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)benzo[d]thiazole-6 -yl)oxazolidin-5-yl)methyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000054
(E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) prepared in Reference Example T025 A mixture of benzo[d]thiazol-6-yl)amino)propyl 4-methylbenzenesulfonate (375mg) and paraformaldehyde (63mg) in acetonitrile (80ml) was stirred at 60°C for 0.5 hours, then paraformaldehyde (42mg). was added, stirred at 60° C. for 0.5 h, cooled to room temperature, passed through a short silica gel column, eluted with ethyl acetate, and concentrated under reduced pressure. The residue was purified by silica gel column chromatography (hexane/ethyl acetate) to give the title compound (166 mg) as a yellow solid.
1 H NMR (300 MHz, DMSO-d 6 ) δ 2.42 (3H, s), 2.84 (3H, d, J = 4.8 Hz), 3.19 (1H, dd, J = 9.2, 6 .5Hz), 3.55 (1H, dd, J = 9.4, 7.3Hz), 4.19 (1H, dd, J = 11.0, 6.1Hz), 4.24 (1H, dd, J = 10.8, 3.5Hz), 4.49-4.61 (1H, m), 4.84 (1H, d, J = 2.8Hz), 4.89 (1H, d, J = 2 .8Hz), 6.77 (1H, dd, J = 9.1, 2.4Hz), 6.85 (1H, d, J = 16.1Hz), 7.13 (1H, d, J = 2.4Hz). 4 Hz), 7.25 (1 H, d, J = 16.1 Hz), 7.48 (2 H, d, J = 7.9 Hz), 7.64 (1 H, q, J = 4.6 Hz), 7. 77-7.85 (3H,m), 7.94 (1H,d,J=1.5Hz), 8.20 (1H,d,J=1.4Hz).
 参考例E001:ジ-tert-ブチル (5-ヨードピラジン-2-イル)-2-イミドジカルボナートの製造
Figure JPOXMLDOC01-appb-C000055
 5-ヨードピラジン-2-アミン(CAS[886860-50-0])(2.86g)のTHF(100ml)溶液に、二炭酸ジ-tert-ブチル(7.06g)及びN,N-ジメチルピリジン-4-アミン(0.316g)を室温で加え室温にて3日間攪拌させた。反応溶液に水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、水及び飽和食塩水で洗浄し、無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、40-60%酢酸エチル/n-ヘプタン)で精製して標題化合物(4.8375g)を得た。
H NMR(400MHz,CDCl)δ(ppm):8.65-8.70(m,1H),8.35-8.39(m,1H),1.43(s,18H).
MS(ESI)m/z:422[M+H]
Reference Example E001: Preparation of di-tert-butyl (5-iodopyrazin-2-yl)-2-imidodicarbonate
Figure JPOXMLDOC01-appb-C000055
To a solution of 5-iodopyrazin-2-amine (CAS[886860-50-0]) (2.86 g) in THF (100 ml) was added di-tert-butyl dicarbonate (7.06 g) and N,N-dimethylpyridine. -4-amine (0.316 g) was added at room temperature and stirred at room temperature for 3 days. Water and ethyl acetate were added to the reaction solution, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate and filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 40-60% ethyl acetate/n-heptane) to give the title compound (4.8375g).
1 H NMR (400 MHz, CDCl 3 ) δ (ppm): 8.65-8.70 (m, 1H), 8.35-8.39 (m, 1H), 1.43 (s, 18H).
MS (ESI) m/z: 422 [M+H] <+> .
 参考例E002:tert-ブチル(5-ヨードピラジン-2-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000056
 参考例E001で製造したジ-tert-ブチル 5-ヨードピラジン-2-イル)-2-イミドジカルボナート(4.8375g)のメタノール(50ml)溶液に炭酸カリウム(4.76g)を室温で加え、混合物を室温で6時間攪拌させたのち、70℃にて1時間攪拌させた。反応溶液を室温に冷却後、水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、水及び飽和食塩水で洗浄し、硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去し標題化合物(3.5023g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.54(s,9H),3.84(s,1H),8.44(d,1H,J=1.8Hz),9.12(d,1H,J=1.4Hz).
MS(ESI)m/z:322[M+H]
Reference Example E002: Preparation of tert-butyl (5-iodopyrazin-2-yl) carbamate
Figure JPOXMLDOC01-appb-C000056
Potassium carbonate (4.76 g) was added to a methanol (50 ml) solution of di-tert-butyl 5-iodopyrazin-2-yl)-2-imidodicarbonate (4.8375 g) prepared in Reference Example E001 at room temperature. The mixture was stirred at room temperature for 6 hours and then at 70° C. for 1 hour. After cooling the reaction solution to room temperature, water and ethyl acetate were added, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with water and saturated brine, dried over sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure to give the title compound (3.5023 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.54 (s, 9H), 3.84 (s, 1H), 8.44 (d, 1H, J=1.8Hz), 9. 12 (d, 1H, J=1.4Hz).
MS (ESI) m/z: 322 [M+H] <+> .
 参考例E003:tert-ブチル(5-ヨードピラジン-2-イル)(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000057
 参考例E002で製造したtert-ブチル(5-ヨードピラジン-2-イル)カルバマート(3.5023g)のDMF(55ml)溶液に、炭酸セシウム(7.11g)及びヨードメタン(1.018ml)を0℃で加えた。混合物を室温にて19時間攪拌させた。反応溶液に水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、水及び飽和食塩水で洗浄し、無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-30%酢酸エチル/n-ヘプタン)で精製して標題化合物(3.2794g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.53(s,9H),3.34(s,3H),8.51(d,1H,J=1.4Hz),8.93(d,1H,J=1.4Hz).
MS(ESI)m/z:336[M+H]
Reference Example E003: Preparation of tert-butyl (5-iodopyrazin-2-yl) (methyl) carbamate
Figure JPOXMLDOC01-appb-C000057
Cesium carbonate (7.11 g) and iodomethane (1.018 ml) were added to a solution of tert-butyl (5-iodopyrazin-2-yl)carbamate (3.5023 g) prepared in Reference Example E002 in DMF (55 ml) at 0°C. added with The mixture was allowed to stir at room temperature for 19 hours. Water and ethyl acetate were added to the reaction solution, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, washed with water and saturated brine, dried over anhydrous sodium sulfate and filtered, and the filtrate was evaporated under reduced pressure. The obtained residue was purified by column chromatography (silica gel, 0-30% ethyl acetate/n-heptane) to give the title compound (3.2794 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.53 (s, 9H), 3.34 (s, 3H), 8.51 (d, 1H, J=1.4 Hz), 8. 93 (d, 1H, J=1.4Hz).
MS (ESI) m/z: 336 [M+H] <+> .
 参考例E004:tert-ブチル(5-(3-ヒドロキシプロプ-1-イン-1-イル)ピラジン-2-イル)(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000058
 参考例E003で製造したtert-ブチル(5-ヨードピラジン-2-イル)(メチル)カルバマート(3.2794g)のトリエチルアミン(9.55ml)懸濁液に、プロパルギルアルコール(1.129ml)、ヨウ化銅(I)(0.224g)及びビス(トリフェニルホスフィン)パラジウム(II)クロリド(0.137g)を加え、窒素雰囲気下中、室温にて90分間攪拌させた。反応溶液に水を加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、30-50%酢酸エチル/n-ヘプタン)で精製して標題化合物(2.1609g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.52(s,9H),3.36(s,3H),4.52(s,2H),8.37(d,1H,J=1.4Hz),9.06(s,1H).MS(ESI)m/z:264[M+H]
Reference Example E004: Preparation of tert-butyl (5-(3-hydroxyprop-1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate
Figure JPOXMLDOC01-appb-C000058
To a suspension of tert-butyl (5-iodopyrazin-2-yl)(methyl)carbamate (3.2794 g) prepared in Reference Example E003 in triethylamine (9.55 ml), propargyl alcohol (1.129 ml), iodide Copper(I) (0.224 g) and bis(triphenylphosphine)palladium(II) chloride (0.137 g) were added and allowed to stir at room temperature for 90 minutes under a nitrogen atmosphere. Water was added to the reaction solution, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous magnesium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The obtained residue was purified by column chromatography (silica gel, 30-50% ethyl acetate/n-heptane) to give the title compound (2.1609 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.52 (s, 9H), 3.36 (s, 3H), 4.52 (s, 2H), 8.37 (d, 1H, J=1.4 Hz), 9.06 (s, 1H). MS (ESI) m/z: 264 [M+H] <+> .
 参考例E005:tert-ブチル メチル(5-(3-オキソプロプ-1-イン-1-イル)ピラジン-2-イル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000059
 塩化オキサリル(1.409ml)のジクロロメタン(40ml)溶液にDMSO(1.747ml)のジクロロメタン(10ml)溶液を-78℃でゆっくりと加え、5分間攪拌させた。反応溶液に、参考例E004で製造したtert-ブチル(5-(3-ヒドロキシプロプ-1-イン-1-イル)ピラジン-2-イル)(メチル)カルバマート(2.1609g)のジクロロメタン(20ml)溶液を-78℃でゆっくりと加え、-78℃にて1時間攪拌させた。反応溶液に-78℃でトリエチルアミン(6.86ml)を加え、-78℃で30分間撹拌させた。反応溶液を室温に昇温し、飽和塩化アンモニウム水溶液及び飽和食塩水を加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、30-50%酢酸エチル/n-ヘプタン)で精製して標題化合物(1.4440g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.49(d,9H,J=1.8Hz),3.34-3.37(m,3H),8.49(d,1H,J=1.4Hz),9.23(d,1H,J=1.4Hz),9.36-9.40(m,1H).
MS(ESI)m/z:262[M+H]
Reference Example E005: Preparation of tert-butyl methyl (5-(3-oxoprop-1-yn-1-yl)pyrazin-2-yl)carbamate
Figure JPOXMLDOC01-appb-C000059
A solution of oxalyl chloride (1.409 ml) in dichloromethane (40 ml) was slowly added with a solution of DMSO (1.747 ml) in dichloromethane (10 ml) at -78°C and stirred for 5 minutes. To the reaction solution was added tert-butyl (5-(3-hydroxyprop-1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate (2.1609 g) prepared in Reference Example E004 in dichloromethane (20 ml). The solution was added slowly at -78°C and allowed to stir at -78°C for 1 hour. Triethylamine (6.86 ml) was added to the reaction solution at -78°C and stirred at -78°C for 30 minutes. The reaction solution was warmed to room temperature, saturated aqueous ammonium chloride solution and saturated brine were added, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 30-50% ethyl acetate/n-heptane) to give the title compound (1.4440 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.49 (d, 9H, J = 1.8 Hz), 3.34-3.37 (m, 3H), 8.49 (d, 1H , J=1.4 Hz), 9.23 (d, 1H, J=1.4 Hz), 9.36-9.40 (m, 1H).
MS (ESI) m/z: 262 [M+H] <+> .
 参考例E006:5-クロロ-2-メチルチアゾロ[5,4-b]ピリジンの製造
Figure JPOXMLDOC01-appb-C000060
 炭酸ナトリウム(7.26g)、3-アミノ-2,6ジクロロピリジン(CAS:62476-56-6,4.29g)及びチオホスゲン(2.62ml)をジクロロメタン(50ml)中に懸濁させ、混合物を室温にて2日間攪拌させた。不溶物をセライト(登録商標)ろ過し、ジクロロメタンで洗浄し、ろ液を減圧留去することで2,6-ジクロロ-3-イソチオシアネートピリジン中間体を得た。これをTHF(12.5ml)に溶解させ、メチルマグネシウムクロリドの3M THF溶液(12.29ml)に窒素雰囲気下で0℃にてゆっくりと滴下した。混合物を0℃にて2時間撹拌させた。混合物に0℃にてゆっくりと飽和塩化アンモニウム水溶液を加えた後、室温へと昇温した。酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去することでN-(2,6-ジクロロピリジン-3-イル)エタンチオアミド中間体を得た。これをDMF(25ml)に溶解させ、炭酸ナトリウム(4.19g)を加えた。混合物を窒素雰囲気下、90℃にて1時間攪拌させた。混合物を氷水へと注ぎ、30分間強く撹拌させた。生じた固体をろ取し、水で洗浄した後、減圧乾燥することで標題化合物(4.84g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):2.84(s,3H),7.39(d,J=8.6Hz,1H),8.09(d,J=8.6Hz,1H).
MS(ESI)m/z:185[M+H]
Reference Example E006: Preparation of 5-chloro-2-methylthiazolo[5,4-b]pyridine
Figure JPOXMLDOC01-appb-C000060
Sodium carbonate (7.26 g), 3-amino-2,6-dichloropyridine (CAS: 62476-56-6, 4.29 g) and thiophosgene (2.62 ml) were suspended in dichloromethane (50 ml) and the mixture was It was allowed to stir at room temperature for 2 days. Insoluble matter was filtered through Celite (registered trademark), washed with dichloromethane, and the filtrate was distilled off under reduced pressure to obtain a 2,6-dichloro-3-isothiocyanatopyridine intermediate. This was dissolved in THF (12.5 ml) and slowly added dropwise to a 3M THF solution of methylmagnesium chloride (12.29 ml) at 0° C. under a nitrogen atmosphere. The mixture was allowed to stir at 0° C. for 2 hours. A saturated ammonium chloride aqueous solution was slowly added to the mixture at 0° C., and then the temperature was raised to room temperature. Ethyl acetate was added and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure to give the N-(2,6-dichloropyridin-3-yl)ethanethioamide intermediate. This was dissolved in DMF (25 ml) and sodium carbonate (4.19 g) was added. The mixture was allowed to stir at 90° C. for 1 hour under a nitrogen atmosphere. The mixture was poured into ice water and allowed to stir vigorously for 30 minutes. The resulting solid was collected by filtration, washed with water, and dried under reduced pressure to obtain the title compound (4.84 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 2.84 (s, 3H), 7.39 (d, J = 8.6 Hz, 1H), 8.09 (d, J = 8.6 Hz , 1H).
MS (ESI) m/z: 185 [M+H] <+> .
 参考例E007:2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジンの製造1
Figure JPOXMLDOC01-appb-C000061
 参考例E006で製造した5-クロロ-2-メチルチアゾロ[5,4-b]ピリジン(CAS:109202-21-3,500mg)のアルファ,アルファ,アルファ-トリフルオロトルエン(18ml)溶液に、N-ブロモスクシンイミド(723mg)及び2,2’-アゾビス(イソブチロニトリル)(89mg)を加え、混合物を80℃にて2時間攪拌させた。混合物に2,2’-アゾビス(イソブチロニトリル)(35.6mg)を追加し、混合物を80℃にて2時間攪拌させた。混合物を室温に冷却後、水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-30%酢酸エチル/n-ヘプタン)で精製して標題化合物(319mg)を得た。H-NMR(400MHz,CDCl)δ(ppm):4.76(d,J=2.3Hz,2H),7.40-7.47(m,1H),8.10-8.23(m,1H).
MS(ESI)m/z:263,265[M+H]
Reference Example E007: Preparation of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine 1
Figure JPOXMLDOC01-appb-C000061
In a solution of 5-chloro-2-methylthiazolo[5,4-b]pyridine (CAS: 109202-21-3,500 mg) prepared in Reference Example E006 in alpha, alpha, alpha-trifluorotoluene (18 ml), N- Bromosuccinimide (723 mg) and 2,2′-azobis(isobutyronitrile) (89 mg) were added and the mixture was allowed to stir at 80° C. for 2 hours. 2,2′-Azobis(isobutyronitrile) (35.6 mg) was added to the mixture and the mixture was stirred at 80° C. for 2 hours. After cooling the mixture to room temperature, water and ethyl acetate were added and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-30% ethyl acetate/n-heptane) to give the title compound (319 mg). 1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 4.76 (d, J = 2.3 Hz, 2H), 7.40-7.47 (m, 1H), 8.10-8.23 (m, 1H).
MS (ESI) m/z: 263,265 [M+H] <+> .
 参考例E008:ジエチル((5-クロロチアゾロ[5,4-b]ピリジン-2-イル)メチル)ホスホネートの製造
Figure JPOXMLDOC01-appb-C000062
 参考例E007で製造した2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジン(319mg)及びトリエチルホスファイト(15ml)の混合物を100℃にて2時間攪拌させた。混合物を室温に冷却後、減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-100%酢酸エチル/n-ヘプタン,5%メタノール/酢酸エチル)で精製して標題化合物(305mg)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.30(dt,J=0.9,7.0Hz,6H),3.64-3.74(m,2H),4.15(q,J=7.4Hz,4H),7.40(dd,J=1.4,8.6Hz,1H),8.14(dd,J=0.9,8.6Hz,1H).
MS(ESI)m/z:321[M+H]
Reference Example E008: Preparation of diethyl ((5-chlorothiazolo[5,4-b]pyridin-2-yl)methyl)phosphonate
Figure JPOXMLDOC01-appb-C000062
A mixture of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine (319 mg) prepared in Reference Example E007 and triethylphosphite (15 ml) was stirred at 100° C. for 2 hours. After the mixture was cooled to room temperature, it was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-100% ethyl acetate/n-heptane, 5% methanol/ethyl acetate) to give the title compound (305 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.30 (dt, J = 0.9, 7.0 Hz, 6H), 3.64-3.74 (m, 2H), 4.15 (q, J=7.4 Hz, 4 H), 7.40 (dd, J=1.4, 8.6 Hz, 1 H), 8.14 (dd, J=0.9, 8.6 Hz, 1 H).
MS (ESI) m/z: 321 [M+H] <+> .
 参考例E009:ジエチル((5-((3,4-ジメトキシベンジル)オキシ)チアゾロ[5,4-b]ピリジン-2-イル)メチル)ホスホナートの製造
Figure JPOXMLDOC01-appb-C000063
 参考例E008で製造したジエチル((5-クロロチアゾロ[5,4-b]ピリジン-2-イル)メチル)ホスホナート(50mg)、3,4-ジメトキシベンジルアルコール(27.2μl)、(R)-(-)-1-[(S)-2-(ジシクロヘキシルホスフィノ)フェロセニル]エチルジ-tert-ブチルホスフィン(12.97mg)、炭酸セシウム(102mg)及びパラジウム(π-シンナミル)クロライドダイマー(4.04mg)のトルエン(0.45ml)中混合物を窒素雰囲気下中、室温にて20時間攪拌させた。混合物を直接カラムクロマトグラフィー(シリカゲル、0-100%酢酸エチル/n-ヘプタン,5%メタノール/酢酸エチル)で精製して標題化合物(33.1mg)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.32(t,J=7.0Hz,6H),3.61-3.71(m,2H),3.89(d,J=6.3Hz,6H),4.16(q,J=7.5Hz,4H),5.35(s,2H),6.83-6.91(m,2H),7.02(s,2H),8.07(d,J=8.6Hz,1H).
MS(ESI)m/z:453[M+H]
Reference Example E009: Preparation of diethyl ((5-((3,4-dimethoxybenzyl)oxy)thiazolo[5,4-b]pyridin-2-yl)methyl)phosphonate
Figure JPOXMLDOC01-appb-C000063
Diethyl ((5-chlorothiazolo[5,4-b]pyridin-2-yl)methyl)phosphonate (50 mg) prepared in Reference Example E008, 3,4-dimethoxybenzyl alcohol (27.2 μl), (R)-( -)-1-[(S)-2-(dicyclohexylphosphino)ferrocenyl]ethyldi-tert-butylphosphine (12.97 mg), cesium carbonate (102 mg) and palladium (π-cinnamyl) chloride dimer (4.04 mg) in toluene (0.45 ml) was allowed to stir at room temperature for 20 hours under a nitrogen atmosphere. The mixture was purified directly by column chromatography (silica gel, 0-100% ethyl acetate/n-heptane, 5% methanol/ethyl acetate) to give the title compound (33.1 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.32 (t, J = 7.0 Hz, 6H), 3.61-3.71 (m, 2H), 3.89 (d, J = 6.3Hz, 6H), 4.16 (q, J = 7.5Hz, 4H), 5.35 (s, 2H), 6.83-6.91 (m, 2H), 7.02 (s , 2H), 8.07 (d, J=8.6 Hz, 1H).
MS (ESI) m/z: 453 [M+H] <+> .
 参考例E010:tert-ブチル(E)-(5-(4-(5-((3,4-ジメトキシベンジル)オキシ)チアゾロ[5,4-b]ピリジン-2-イル)ブタ-3-エン-1-イン-1-イル)ピラジン-2-イル)(メチル)カルバマートの製造
Figure JPOXMLDOC01-appb-C000064
 参考例E009で製造したジエチル((5-((3,4-ジメトキシベンジル)オキシ)チアゾロ[5,4-b]ピリジン-2-イル)メチル)ホスホネート(33mg)のTHF(1.5ml)溶液に、0℃で水素化ナトリウム(4.38mg,60%オイルディスパージョン)を加え、0℃にて30分間攪拌させた。混合物に0℃で、参考例E005で製造したtert-ブチル メチル(5-(3-オキソプロプ-1-イン-1-イル)ピラジン-2-イル)カルバマート(38.1mg)のTHF(0.75ml)溶液を加え、0℃で1時間撹拌させた。混合物に飽和塩化アンモニウム水溶液を加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-30%酢酸エチル/n-ヘプタン)で精製して標題化合物(28.6mg)を得た。
H-NMR(400MHz,CDCl)δ(ppm):1.55(s,9H),3.39-3.42(m,3H),3.86-3.91(m,6H),5.37(s,2H),6.78(d,J=15.9Hz,1H),6.85-6.92(m,2H),7.00-7.07(m,2H),7.27(d,J=16.3Hz,1H),8.09(d,J=9.1Hz,1H),8.45(d,J=1.4Hz,1H),9.15(d,J=1.4Hz,1H).
MS(ESI)m/z:560[M+H]
Reference Example E010: tert-butyl (E)-(5-(4-(5-((3,4-dimethoxybenzyl)oxy)thiazolo[5,4-b]pyridin-2-yl)but-3-ene Preparation of 1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate
Figure JPOXMLDOC01-appb-C000064
Diethyl ((5-((3,4-dimethoxybenzyl)oxy)thiazolo[5,4-b]pyridin-2-yl)methyl)phosphonate (33mg) prepared in Reference Example E009 in THF (1.5ml) To the solution was added sodium hydride (4.38 mg, 60% oil dispersion) at 0°C and stirred at 0°C for 30 minutes. THF (0.75 ml) of tert-butyl methyl (5-(3-oxoprop-1-yn-1-yl)pyrazin-2-yl)carbamate (38.1 mg) prepared in Reference Example E005 was added to the mixture at 0°C. ) solution and allowed to stir at 0° C. for 1 hour. A saturated aqueous ammonium chloride solution was added to the mixture, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous magnesium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-30% ethyl acetate/n-heptane) to give the title compound (28.6 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 1.55 (s, 9H), 3.39-3.42 (m, 3H), 3.86-3.91 (m, 6H), 5.37 (s, 2H), 6.78 (d, J=15.9Hz, 1H), 6.85-6.92 (m, 2H), 7.00-7.07 (m, 2H), 7.27 (d, J = 16.3Hz, 1H), 8.09 (d, J = 9.1Hz, 1H), 8.45 (d, J = 1.4Hz, 1H), 9.15 (d , J=1.4 Hz, 1H).
MS (ESI) m/z: 560 [M+H] <+> .
 参考例E011:(E)-2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-オールの製造
Figure JPOXMLDOC01-appb-C000065
 参考例E010で製造したtert-ブチル(E)-(5-(4-(5-((3,4-ジメトキシベンジル)オキシ)チアゾロ[5,4-b]ピリジン-2-イル)ブタ-3-エン-1-イン-1-イル)ピラジン-2-イル)(メチル)カルバマート(28.6mg)のジクロロメタン(0.8ml)溶液に、0℃でトリフルオロ酢酸(0.5ml)を加え、室温にて4時間攪拌させた。混合物を減圧して未精製の標題化合物(32.8mg)を得た。
H-NMR(400MHz,DMSO-d)δ(ppm):2.79(s,3H),6.71(brd,J=9.1Hz,1H),6.86(brd,J=16.3Hz,1H),7.19(d,J=15.9Hz,1H),7.91(s,1H),8.09(brd,J=9.1,1H),8.14(s,1H).
MS(ESI)m/z:310[M+H]
Reference Example E011: (E)-2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4-b]pyridine- Production of 5-ol
Figure JPOXMLDOC01-appb-C000065
tert-Butyl (E)-(5-(4-(5-((3,4-dimethoxybenzyl)oxy)thiazolo[5,4-b]pyridin-2-yl)but-3 prepared in Reference Example E010 -en-1-yn-1-yl)pyrazin-2-yl)(methyl)carbamate (28.6mg) in dichloromethane (0.8ml) was added with trifluoroacetic acid (0.5ml) at 0°C, The mixture was stirred at room temperature for 4 hours. The mixture was decompressed to give the crude title compound (32.8mg).
1 H-NMR (400 MHz, DMSO-d 6 ) δ (ppm): 2.79 (s, 3H), 6.71 (brd, J = 9.1 Hz, 1H), 6.86 (brd, J = 16 .3 Hz, 1 H), 7.19 (d, J = 15.9 Hz, 1 H), 7.91 (s, 1 H), 8.09 (brd, J = 9.1, 1 H), 8.14 (s , 1H).
MS (ESI) m/z: 310 [M+H] <+> .
 参考例E012:(E)-2-((tert-ブチルジメチルシリル)オキシ)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000066
 参考例E011で製造した未精製の(E)-2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-オール(1.81g)、WO2022/45093号の参考例15の製法により製造した2-((tert-ブチルジメチルシリル)オキシ)-3-ヒドロキシプロピル 4-メチルベンゼンスルホナート(2.63g)及びトリフェニルホスフィン(3.06g)のTHF(29ml)溶液に、アゾジカルボン酸ジイソプロピル(40%トルエン溶液、6.14ml)を0℃で加え、0℃で5時間撹拌した。混合物に水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧下で濃縮した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-50%酢酸エチル/n-ヘプタン)で精製して標題化合物(3.82g)を得た。
MS(ESI)m/z:652[M+H]
Reference Example E012: (E)-2-((tert-butyldimethylsilyl)oxy)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-ene-3 -yn-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propyl 4-methylbenzenesulfonate preparation
Figure JPOXMLDOC01-appb-C000066
Crude (E)-2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4 prepared in Reference Example E011 -b]pyridin-5-ol (1.81 g), 2-((tert-butyldimethylsilyl)oxy)-3-hydroxypropyl 4-methylbenzenesulfonate prepared by the method of Reference Example 15 of WO2022/45093 (2.63 g) and triphenylphosphine (3.06 g) in THF (29 ml) was added diisopropyl azodicarboxylate (40% toluene solution, 6.14 ml) at 0°C and stirred at 0°C for 5 hours. Water and ethyl acetate were added to the mixture, and the aqueous layer was extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-50% ethyl acetate/n-heptane) to give the title compound (3.82 g).
MS (ESI) m/z: 652 [M+H] <+> .
 参考例E013:(E)-2-ヒドロキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000067
 参考例E012で製造した(E)-2-((tert-ブチルジメチルシリル)オキシ)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロピル 4-メチルベンゼンスルホナート(3.42g)のTHF(50ml)溶液に、酢酸(6.01ml)及びテトラ-n-ブチルアンモニウムフルオリド(1M THF溶液,26.2ml)を0℃で加えた。その混合物を室温で18時間撹拌した後、カラムクロマトグラフィー(ODSゲル、0-80%アセトニトリル(0.1%ギ酸含有)/水(0.1%ギ酸含有))で精製し標題化合物(269mg)を得た。
H-NMR(400MHz,CDCl)δ(ppm):2.42(s,3H),3.04(d,J=3.2Hz,3H),4.12-4.30(m,3H),4.46(d,J=5.0Hz,2H),5.10(brs,1H),6.75-6.86(m,2H),7.19(d,J=15.9Hz,1H),7.32(d,J=8.2Hz,2H),7.80(d,J=8.6Hz,2H),7.92(d,J=1.8Hz,1H),8.09(d,J=8.6Hz,1H),8.22(d,J=1.4Hz,1H).
MS(ESI)m/z:538[M+H]
Reference Example E013: (E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo [ Preparation of 5,4-b]pyridin-5-yl)oxy)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000067
(E)-2-((tert-butyldimethylsilyl)oxy)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-ene prepared in Reference Example E012) -3-yn-1-yl)thiazolo[5,4-b]pyridin-5-yl)oxy)propyl 4-methylbenzenesulfonate (3.42 g) in THF (50 ml) was dissolved in acetic acid (6.01 ml). ) and tetra-n-butylammonium fluoride (1M THF solution, 26.2 ml) were added at 0°C. The mixture was stirred at room temperature for 18 hours and then purified by column chromatography (ODS gel, 0-80% acetonitrile (containing 0.1% formic acid)/water (containing 0.1% formic acid)) to give the title compound (269 mg). got
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 2.42 (s, 3H), 3.04 (d, J = 3.2 Hz, 3H), 4.12-4.30 (m, 3H ), 4.46 (d, J = 5.0 Hz, 2H), 5.10 (brs, 1H), 6.75-6.86 (m, 2H), 7.19 (d, J = 15.9 Hz , 1H), 7.32 (d, J = 8.2 Hz, 2H), 7.80 (d, J = 8.6 Hz, 2H), 7.92 (d, J = 1.8 Hz, 1H), 8 .09 (d, J=8.6 Hz, 1 H), 8.22 (d, J=1.4 Hz, 1 H).
MS (ESI) m/z: 538 [M+H] <+> .
 参考例E014:2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジン及び5-クロロ-2-(ジブロモメチル)チアゾロ[5,4-b]ピリジンの製造
Figure JPOXMLDOC01-appb-C000068
 参考例E006で製造した5-クロロ-2-メチルチアゾロ[5,4-b]ピリジン(CAS:109202-21-3,3g)のアルファ,アルファ,アルファ-トリフルオロトルエン(108ml)溶液にN-ブロモスクシンイミド(4.34g)及び2,2’-アゾビス(イソブチロニトリル)(534mg)を室温で加え、混合物を95℃にて2時間撹拌させた。混合物に2,2’-アゾビス(イソブチロニトリル)(213mg)を追加し、混合物を95℃にて2時間撹拌させた。混合物に、水及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を無水硫酸ナトリウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-10%酢酸エチル/n-ヘプタン)で精製して2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジン(534mg)と5-クロロ-2-(ジブロモメチル)チアゾロ[5,4-b]ピリジン(2.88g)とを分離して得た。
2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジン:
H-NMR(400MHz,CDCl)δ(ppm):4.76(s,2H),7.46(d,J=8.6Hz,1H),8.18(d,J=8.6Hz,1H).
MS(ESI)m/z:263,265[M+H]
5-クロロ-2-(ジブロモメチル)チアゾロ[5,4-b]ピリジン:
H-NMR(400MHz,CDCl)δ(ppm):6.86(s,1H),7.49(d,J=8.6Hz,1H),8.19(d,J=8.6Hz,1H).
MS(ESI)m/z:343,345[M+H]
Reference Example E014: Preparation of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine and 5-chloro-2-(dibromomethyl)thiazolo[5,4-b]pyridine
Figure JPOXMLDOC01-appb-C000068
N-bromo Succinimide (4.34 g) and 2,2′-azobis(isobutyronitrile) (534 mg) were added at room temperature and the mixture was allowed to stir at 95° C. for 2 hours. 2,2′-azobis(isobutyronitrile) (213 mg) was added to the mixture and the mixture was allowed to stir at 95° C. for 2 hours. Water and ethyl acetate were added to the mixture, and the aqueous layer was extracted with ethyl acetate. The organic layer was dried over anhydrous sodium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-10% ethyl acetate/n-heptane) to give 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine (534 mg) and 5-chloro -2-(Dibromomethyl)thiazolo[5,4-b]pyridine (2.88 g) was isolated.
2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine:
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 4.76 (s, 2H), 7.46 (d, J = 8.6 Hz, 1H), 8.18 (d, J = 8.6 Hz , 1H).
MS (ESI) m/z: 263,265 [M+H] +
5-chloro-2-(dibromomethyl)thiazolo[5,4-b]pyridine:
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 6.86 (s, 1H), 7.49 (d, J = 8.6 Hz, 1H), 8.19 (d, J = 8.6 Hz , 1H).
MS (ESI) m/z: 343, 345 [M+H] <+> .
 参考例E015:2-(ブロモメチル)-5-クロロチアゾロ[5,4-b]ピリジンの製造2
Figure JPOXMLDOC01-appb-C000069
 参考例E014で製造した5-クロロ-2-(ジブロモメチル)チアゾロ[5,4-b]ピリジン(2.33g)のTHF(14ml)溶液に0℃でジイソプロピルエチルアミン(1.66ml)及び亜りん酸ジエチル(1.58ml)を加え、0℃で1時間撹拌させた。混合物に室温で亜りん酸ジエチル(0.526ml)を加え、30分間撹拌させた。混合物に酢酸エチル、水及び飽和塩化アンモニウム水溶液を加え、水層を酢酸エチルで抽出した。有機層を水及び飽和食塩水で洗浄後、少量のシリカゲルを通してろ過し、無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(シリカゲル、3-13%酢酸エチル/n-ヘプタン)で精製して標題化合物(1.58g)を得た。
H-NMR(400MHz,CDCl)δ(ppm):4.76(s,2H),7.46(d,J=8.6Hz,1H),8.17(d,J=8.6Hz,1H).
MS(ESI)m/z:263,265[M+H]
Reference Example E015: Preparation of 2-(bromomethyl)-5-chlorothiazolo[5,4-b]pyridine 2
Figure JPOXMLDOC01-appb-C000069
Diisopropylethylamine (1.66 ml) and phosphorus Diethyl acid (1.58 ml) was added and stirred at 0° C. for 1 hour. Diethyl phosphite (0.526 ml) was added to the mixture at room temperature and stirred for 30 minutes. Ethyl acetate, water and saturated aqueous ammonium chloride solution were added to the mixture, and the aqueous layer was extracted with ethyl acetate. The organic layer was washed with water and saturated brine, filtered through a small amount of silica gel, dried over anhydrous magnesium sulfate and filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 3-13% ethyl acetate/n-heptane) to give the title compound (1.58 g).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 4.76 (s, 2H), 7.46 (d, J = 8.6 Hz, 1H), 8.17 (d, J = 8.6 Hz , 1H).
MS (ESI) m/z: 263,265 [M+H] <+> .
 実施例5:(E)-1-フルオロ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロパン-2-オール(化合物(III)。以下、「SPAL-E-17」ともいう)の製造1
Figure JPOXMLDOC01-appb-C000070
 参考例E011で製造した未精製の(E)-2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-オール(32.8mg)及び炭酸カリウム(14.2mg)のDMF(500μl)中混合物に、2-(フルオロメチル)オキシラン(CAS:503-09-3,12.8μl)を室温で加えた。混合物を80℃にて2時間攪拌させた。混合物を室温に冷却後、飽和塩化アンモニウム水溶液及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を合わせ、無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧留去した。得られた残渣をカラムクロマトグラフィー(ODSゲル、15-50%アセトニトリル(0.1%ギ酸含有)/水(0.1%ギ酸含有))で精製して標題化合物(2.6mg)を得た。H-NMR(400MHz,CDCl)δ(ppm):3.03(d,J=5.4Hz,3H),3.85(d,J=5.0Hz,1H),4.23-4.35(m,1H),4.48-4.58(m,3H),4.59-4.68(m,1H),4.90(brd,J=5.0Hz,1H),6.78(d,J=15.9Hz,1H),6.90(d,J=9.1Hz,1H),7.18(d,J=15.9Hz,1H),7.89(d,J=1.4Hz,1H),8.10(d,J=9.1Hz,1H),8.21(d,J=1.4Hz,1H).
MS(ESI)m/z:386[M+H]
Example 5: (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo [ Preparation 1 of 5,4-b]pyridin-5-yl)oxy)propan-2-ol (compound (III), hereinafter also referred to as “SPAL-E-17”)
Figure JPOXMLDOC01-appb-C000070
Crude (E)-2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4 prepared in Reference Example E011 2-(fluoromethyl)oxirane (CAS: 503-09-3, 12.8 μl) to a mixture of b]pyridin-5-ol (32.8 mg) and potassium carbonate (14.2 mg) in DMF (500 μl) was added at room temperature. The mixture was allowed to stir at 80° C. for 2 hours. After cooling the mixture to room temperature, saturated aqueous ammonium chloride solution and ethyl acetate were added, and the aqueous layer was extracted with ethyl acetate. The organic layers were combined, dried over anhydrous magnesium sulfate, filtered, and the filtrate was evaporated under reduced pressure. The resulting residue was purified by column chromatography (ODS gel, 15-50% acetonitrile (containing 0.1% formic acid)/water (containing 0.1% formic acid)) to give the title compound (2.6 mg). . 1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 3.03 (d, J = 5.4 Hz, 3H), 3.85 (d, J = 5.0 Hz, 1H), 4.23-4 .35 (m, 1H), 4.48-4.58 (m, 3H), 4.59-4.68 (m, 1H), 4.90 (brd, J=5.0Hz, 1H), 6 .78 (d, J = 15.9 Hz, 1 H), 6.90 (d, J = 9.1 Hz, 1 H), 7.18 (d, J = 15.9 Hz, 1 H), 7.89 (d, J=1.4 Hz, 1 H), 8.10 (d, J=9.1 Hz, 1 H), 8.21 (d, J=1.4 Hz, 1 H).
MS (ESI) m/z: 386 [M+H] <+> .
 実施例6:(E)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)-2-((テトラヒドロ-2H-ピラン-2-イル)オキシ)プロピル 4-メチルベンゼンスルホナートの製造
Figure JPOXMLDOC01-appb-C000071
 参考例E013で製造した(E)-2-ヒドロキシ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロピル 4-メチルベンゼンスルホナート(269mg)のジクロロメタン(5ml)溶液に、3,4-ジヒドロ-2H-ピラン(0.091ml)及びp-トルエンスルホン酸一水和物(47.7mg)を0℃で加えた。その混合物を室温で1時間撹拌した後、3,4-ジヒドロ-2H-ピラン(0.046ml)を0℃で加えた。その混合物を室温で30分間撹拌した後、0℃で飽和炭酸水素ナトリウム水溶液及び酢酸エチルを加え、水層を酢酸エチルで抽出した。有機層を無水硫酸マグネシウムで乾燥し、ろ過し、ろ液を減圧下で濃縮した。得られた残渣をカラムクロマトグラフィー(シリカゲル、0-100%酢酸エチル/n-ヘプタン)で精製して標題化合物(57.5mg)を得た。
MS(ESI)m/z:622[M+H]
Example 6: (E)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo[5,4- Preparation of b]pyridin-5-yl)oxy)-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate
Figure JPOXMLDOC01-appb-C000071
(E)-2-hydroxy-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) prepared in Reference Example E013 To a solution of thiazolo[5,4-b]pyridin-5-yl)oxy)propyl 4-methylbenzenesulfonate (269 mg) in dichloromethane (5 ml) was added 3,4-dihydro-2H-pyran (0.091 ml) and p - Toluenesulfonic acid monohydrate (47.7 mg) was added at 0°C. After the mixture was stirred at room temperature for 1 hour, 3,4-dihydro-2H-pyran (0.046 ml) was added at 0°C. After the mixture was stirred at room temperature for 30 minutes, saturated aqueous sodium hydrogencarbonate solution and ethyl acetate were added at 0° C., and the aqueous layer was extracted with ethyl acetate. The organic layer was dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by column chromatography (silica gel, 0-100% ethyl acetate/n-heptane) to give the title compound (57.5 mg).
MS (ESI) m/z: 622 [M+H] <+> .
 実施例7:(E)-1-フルオロ-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)プロパン-2-オール(化合物III)の製造2
Figure JPOXMLDOC01-appb-C000072
 フッ化カリウム(2.8mg)及びヨウ化テトラブチルアンモニウム(1.49mg)の水(0.50ml)溶液に、4,7,13,16,21,24-ヘキサオキサ-1,10-ジアザビシクロ[8.8.8]ヘキサコサン(Kryptofix(登録商標)222)(18.2mg)のアセトニトリル(0.50ml)溶液を室温で加え、その混合物を40℃、減圧下で濃縮した。得られた残渣にアセトニトリル(1.0ml)を加え、40℃、減圧下で濃縮した。再度、得られた残渣にアセトニトリル(1.0ml)を加え、40℃、減圧下で濃縮した。得られた残渣にトルエン(1.0ml)を加え、40℃、減圧下で濃縮した。得られた残渣にアセトニトリル(1.0ml)を加え、40℃、減圧下で濃縮した。得られた残渣を100℃で10分間、減圧下で乾燥した。得られた残渣に実施例6で製造した(E)-3-((2-(4-(5-(メチルアミノ)ピラジン-2-イル)ブタ-1-エン-3-イン-1-イル)チアゾロ[5,4-b]ピリジン-5-イル)オキシ)-2-((テトラヒドロ-2H-ピラン-2-イル)オキシ)プロピル 4-メチルベンゼンスルホナート(5.0mg)のDMSO(0.60ml)溶液を60℃で加え、60℃で90分間撹拌した。反応液を室温まで冷却した後、水(0.30ml)及びトリフルオロ酢酸(0.70ml)を加え、5分間室温で撹拌した。その混合物をカラムクロマトグラフィー(ODSゲル、0-40%アセトニトリル(0.1%ギ酸含有)/水(0.1%ギ酸含有))で精製して標題化合物(2.1mg)を得た。
H-NMR(400MHz,CDCl)δ(ppm):3.03(d,J=5.0Hz,3H),4.24-4.34(m,2H),4.46-4.58(m,3H),4.63(t,J=4.8Hz,1H),5.03(brd,J=4.1Hz,1H),6.74-6.83(m,1H),6.90(d,J=9.1Hz,1H),7.18(d,J=15.9Hz,1H),7.91(s,1H),8.10(d,J=8.6Hz,1H),8.21(s,1H).
MS(ESI)m/z:386[M+H]
 (試験例)
Example 7: (E)-1-fluoro-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl)thiazolo [ Preparation of 5,4-b]pyridin-5-yl)oxy)propan-2-ol (Compound III) 2
Figure JPOXMLDOC01-appb-C000072
4,7,13,16,21,24-hexaoxa-1,10-diazabicyclo[8 .8.8] A solution of hexacosane (Kryptofix® 222) (18.2 mg) in acetonitrile (0.50 ml) was added at room temperature and the mixture was concentrated under reduced pressure at 40°C. Acetonitrile (1.0 ml) was added to the resulting residue, and the mixture was concentrated at 40° C. under reduced pressure. Acetonitrile (1.0 ml) was added again to the obtained residue, and the mixture was concentrated at 40° C. under reduced pressure. Toluene (1.0 ml) was added to the resulting residue, and the mixture was concentrated under reduced pressure at 40°C. Acetonitrile (1.0 ml) was added to the resulting residue, and the mixture was concentrated at 40° C. under reduced pressure. The resulting residue was dried at 100° C. for 10 minutes under reduced pressure. (E)-3-((2-(4-(5-(methylamino)pyrazin-2-yl)but-1-en-3-yn-1-yl) prepared in Example 6 was added to the resulting residue. ) Thiazolo[5,4-b]pyridin-5-yl)oxy)-2-((tetrahydro-2H-pyran-2-yl)oxy)propyl 4-methylbenzenesulfonate (5.0 mg) in DMSO (0 .60 ml) solution was added at 60° C. and stirred at 60° C. for 90 minutes. After cooling the reaction solution to room temperature, water (0.30 ml) and trifluoroacetic acid (0.70 ml) were added, and the mixture was stirred at room temperature for 5 minutes. The mixture was purified by column chromatography (ODS gel, 0-40% acetonitrile (containing 0.1% formic acid)/water (containing 0.1% formic acid)) to give the title compound (2.1 mg).
1 H-NMR (400 MHz, CDCl 3 ) δ (ppm): 3.03 (d, J = 5.0 Hz, 3H), 4.24-4.34 (m, 2H), 4.46-4.58 (m, 3H), 4.63 (t, J = 4.8Hz, 1H), 5.03 (brd, J = 4.1Hz, 1H), 6.74-6.83 (m, 1H), 6 .90 (d, J = 9.1 Hz, 1 H), 7.18 (d, J = 15.9 Hz, 1 H), 7.91 (s, 1 H), 8.10 (d, J = 8.6 Hz, 1H), 8.21(s, 1H).
MS (ESI) m/z: 386 [M+H] <+> .
(Test example)
 [ヒト脳の光学イメージング]
 (解剖脳組織)
 死後ヒト脳を、レビー小体型認知症(DLB)患者、及び、アルツハイマー病(AD)患者に対して行われた剖検から得た。凍結DLB組織をクリオスタット(HM560、Carl Zeiss)内で20μm厚にスライスした。また、AD脳組織を10%中性緩衝ホルマリンに固定し、パラフィンブロックに埋め込み、6μm厚にスライスした。
[Optical imaging of the human brain]
(dissected brain tissue)
Postmortem human brains were obtained from autopsies performed on dementia with Lewy bodies (DLB) and Alzheimer's disease (AD) patients. Frozen DLB tissue was sliced 20 μm thick in a cryostat (HM560, Carl Zeiss). AD brain tissue was also fixed in 10% neutral buffered formalin, embedded in paraffin blocks, and sliced at 6 μm thickness.
 (インビトロ蛍光顕微鏡測定)
 DLB患者脳扁桃体組織の後固定新鮮凍結切片、及び、脱パラフィンしたAD患者脳上側頭回組織のホルマリン固定パラフィン包埋切片を使用した。30μmol/lの試験化合物と脳切片を50%エタノール溶液中にて30分間室温でインキュベートした。その後切片を50%エタノール溶液で5分間、超純水で3分間、2回洗浄した。封入剤(VECTASHIELD H-1000、Vector Laboratories)を用いて切片を封入後、蛍光顕微鏡(DM4000、Leica)を用いて切片上の病変蓄積領域の画像を取得した、蛍光画像を図1に示す。図1において、矢頭はDLB患者脳のαシヌクレイン凝集体に結合した化合物の蛍光を、矢印はAD患者脳のアミロイドβ凝集体に結合した化合物の蛍光を、アスタリスクはAD患者脳のタウ凝集体に結合した化合物の蛍光を示す。解析ソフトウェア(Image J)を用いて病変領域及び病変非形成領域(バックグラウンド)の蛍光輝度を定量した。結果を図2に示す。なお、試験化合物としては、SPAL-T-83、SPAL-T-95及びSPAL-E-17を用いた。また対照の化合物としては、上述の特許文献2(国際公開第2020/174963号)の合成実施例21に記載された方法により合成した((E)-1-フルオロ-3-(2-(4-(6-(メチルアミノ)ピリジン-3-イル)ブタ-1-エン-3-イニル)ベンゾ[d]チアゾール-6-イルオキシ)プロパン-2-オール(C05-05)を使用した。
(In vitro fluorescence microscope measurement)
Post-fixed fresh frozen sections of DLB patient brain amygdala tissue and formalin-fixed paraffin-embedded sections of deparaffinized AD patient brain superior temporal gyrus tissue were used. 30 μmol/l test compound and brain slices were incubated in a 50% ethanol solution for 30 minutes at room temperature. After that, the sections were washed twice with a 50% ethanol solution for 5 minutes and then with ultrapure water for 3 minutes. After mounting the section using a mounting medium (VECTASHIELD H-1000, Vector Laboratories), an image of the lesion accumulation area on the section was obtained using a fluorescence microscope (DM4000, Leica). A fluorescence image is shown in FIG. In FIG. 1, arrowheads indicate fluorescence of compounds bound to α-synuclein aggregates in DLB patient brains, arrows indicate fluorescence of compounds bound to amyloid β aggregates in AD patient brains, and asterisks indicate tau aggregates in AD patient brains. Fluorescence of bound compound is shown. Analysis software (Image J) was used to quantify the fluorescence intensity of lesion areas and non-lesion forming areas (background). The results are shown in FIG. SPAL-T-83, SPAL-T-95 and SPAL-E-17 were used as test compounds. As a control compound, ((E)-1-fluoro-3-(2-(4 -(6-(methylamino)pyridin-3-yl)but-1-en-3-ynyl)benzo[d]thiazol-6-yloxy)propan-2-ol (C05-05) was used.
 図1及び図2に示すように、SPAL-T-83、SPAL-T-95及びSPAL-E-17はDLBの患者脳で形成されるαシヌクレイン凝集体に結合していることが確認された。また、SPAL-T-83、SPAL-T-95及びSPAL-E-17のαシヌクレイン凝集体への結合性は、ADの患者脳で形成されるタウ又はアミロイドβの凝集体への結合よりも強いことが確認された。さらに、タウ又はアミロイドβ凝集体への結合に対するαシヌクレイン凝集体への結合の比は、C05-05よりも高いことが確認された。 As shown in FIGS. 1 and 2, SPAL-T-83, SPAL-T-95 and SPAL-E-17 were confirmed to bind to α-synuclein aggregates formed in DLB patient brains. . Also, the binding of SPAL-T-83, SPAL-T-95 and SPAL-E-17 to α-synuclein aggregates was significantly higher than to tau or amyloid β aggregates formed in AD patient brains. confirmed to be strong. Furthermore, the ratio of binding to alpha-synuclein aggregates to binding to tau or amyloid beta aggregates was confirmed to be higher than for C05-05.
 [マウス脳の光学イメージング]
 (αシヌクレイン線維接種マウスモデルの作製)
 マウスαシヌクレインをリコンビナントタンパク質として大腸菌で発現させて抽出し、試験管内でインキュベートすると、不溶性の凝集体を形成する。このαシヌクレイン凝集体をマウスの線条体に接種すると、神経回路を経路としてαシヌクレインの凝集体が周囲の領域に伝播し、数か月後には大脳新皮質でαシヌクレイン病変が観察される(Masuda-Suzukake et al. Acta Neuropathol Commun 2, 88, 2014; Shimozawa et al. Acta Neuropathol Commun 5, 12, 2017)。このマウスの脳を摘出し、切片を作製して蛍光染色で解析すると、リン酸化αシヌクレインからなる病変に、本発明の化合物が結合するかどうかを確認することができる。
[Optical Imaging of Mouse Brain]
(Preparation of α-synuclein fiber-inoculated mouse model)
Mouse α-synuclein is expressed as a recombinant protein in E. coli, extracted, and incubated in vitro to form insoluble aggregates. When this α-synuclein aggregate is inoculated into the striatum of mice, the α-synuclein aggregate propagates to surrounding areas via neural circuits, and several months later, α-synuclein lesions are observed in the cerebral neocortex ( Masuda-Suzukake et al. Acta Neuropathol Commun 2, 88, 2014; Shimozawa et al. Acta Neuropathol Commun 5, 12, 2017). The brain of this mouse is excised, sectioned, and analyzed by fluorescent staining to confirm whether the compound of the present invention binds to lesions composed of phosphorylated α-synuclein.
 (αシヌクレイン線維接種マウスモデル作製)
 まず、マウスαシヌクレインをリコンビナントタンパク質として大腸菌で発現させて抽出し、試験管内でインキュベートして、不溶性のαシヌクレイン凝集体を形成した。そして、このαシヌクレイン凝集体をマウスの線条体に接種したところ、神経回路を経路としてαシヌクレインの凝集体が周囲の領域に伝播し、数か月後には大脳新皮質でαシヌクレイン病変が観察された。なお、このαシヌクレイン凝集体のマウスの線条体への接種は、以下の方法で行った。まず、1.5%(v/v)イソフルランで麻酔した9週齢のC57/BL/6オスマウスの頭部の毛を取り除き、頭皮をイソジンで消毒後、キシロカインを塗り、頭皮に切れ込みを入れて頭蓋を露出させた。そして、Bregma 0.05mm、Lateral 2mmの位置の頭蓋にドリルで穴を開け、ガラスピペットを用いて2mmの深さ位置にαシヌクレイン線維溶液(マウスαシヌクレイン線維 4mg/ml in saline)3μlを注入した後、頭皮を戻して縫合した。
(Preparation of α-synuclein fiber-inoculated mouse model)
First, mouse α-synuclein was expressed in E. coli as a recombinant protein, extracted, and incubated in vitro to form insoluble α-synuclein aggregates. When these α-synuclein aggregates were inoculated into the striatum of mice, α-synuclein aggregates propagated to the surrounding area via neural circuits, and several months later, α-synuclein lesions were observed in the cerebral neocortex. was done. Inoculation of this α-synuclein aggregate into the mouse striatum was performed by the following method. First, the head of a 9-week-old C57/BL/6 male mouse anesthetized with 1.5% (v/v) isoflurane was dehaired, the scalp was disinfected with isodine, xylocaine was applied, and the scalp was incised. The skull was exposed. Then, a hole was drilled in the skull at a position of Bregma 0.05 mm and Lateral 2 mm, and 3 μl of α-synuclein fiber solution (mouse α-synuclein fiber 4 mg/ml in saline) was injected at a depth of 2 mm using a glass pipette. After that, the scalp was put back and sutured.
 (インビトロ蛍光顕微鏡測定)
 このαシヌクレイン線維接種マウスの脳を摘出し、切片を作製して蛍光染色で解析した。具体的には、αシヌクレイン線維接種マウス脳切片と30μmol/lの試験化合物を20%エタノール溶液中にて30分間室温でインキュベートした。その後切片を20%エタノール溶液で5分間、超純水で3分間、2回洗浄した。封入剤(VECTASHIELD H-1000)を用いて切片を封入後、蛍光顕微鏡(DM4000)を用いて切片上のαシヌクレイン凝集体蓄積領域の画像を取得した。同一切片をリン酸緩衝液を用いて洗浄後、抗原性賦活化のためにオートクレーブで処理した。抗リン酸化αシヌクレインモノクローナル抗体(pS129、abcam、ab59264)(1:1000)による免疫組織化学染色を行い、封入剤(VECTASHIELD H-1000)を用いて切片を封入後、蛍光顕微鏡(DM4000)を用いて上記と同一領域の画像を取得した。結果を図3に示す。図3の(a)、(b)及び(c)はそれぞれSPAL-T-83、SPAL-T-95及びSPAL-E-17についての結果を示し、図3の(a)、(b)及び(c)において、右側が抗リン酸化αシヌクレイン抗体染色、左側がSPAL-T-83、SPAL-T-95及びSPAL-E-17蛍光染色である。図3において、矢頭はαシヌクレイン線維接種マウス脳のリン酸化αシヌクレインからなる病変、及びそれらに結合した化合物の蛍光を示している。
(In vitro fluorescence microscope measurement)
The brain of this α-synuclein fiber-inoculated mouse was excised, sectioned, and analyzed by fluorescent staining. Specifically, α-synuclein fiber-inoculated mouse brain sections and 30 μmol/l test compound were incubated in a 20% ethanol solution for 30 minutes at room temperature. After that, the sections were washed twice with a 20% ethanol solution for 5 minutes and then with ultrapure water for 3 minutes. After mounting the sections using a mounting medium (VECTASHIELD H-1000), images of α-synuclein aggregate accumulation areas on the sections were obtained using a fluorescence microscope (DM4000). The same piece was washed with phosphate buffer and then autoclaved for antigen retrieval. Immunohistochemical staining with an anti-phosphorylated α-synuclein monoclonal antibody (pS129, abcam, ab59264) (1:1000) was performed, and after mounting the sections using a mounting medium (VECTASHIELD H-1000), a fluorescence microscope (DM4000) was used. An image of the same region as above was acquired by The results are shown in FIG. (a), (b) and (c) of FIG. 3 show the results for SPAL-T-83, SPAL-T-95 and SPAL-E-17, respectively, and (a), (b) and (c) of FIG. In (c), the right side is anti-phosphorylated α-synuclein antibody staining, and the left side is SPAL-T-83, SPAL-T-95 and SPAL-E-17 fluorescent staining. In FIG. 3, arrowheads indicate lesions composed of phosphorylated α-synuclein in α-synuclein fiber-inoculated mouse brain and fluorescence of compounds bound to them.
 図3の結果から、リン酸化αシヌクレインからなる病変に、SPAL-T-83、SPAL-T-95及びSPAL-E-17が結合することが示された。 The results in Figure 3 showed that SPAL-T-83, SPAL-T-95 and SPAL-E-17 bound to lesions composed of phosphorylated α-synuclein.
 [インビボ二光子レーザースキャニング蛍光顕微鏡検査]
 αシヌクレイン線維溶液注入4か月以降のモデルマウスを1.5%(v/v)イソフルランで麻酔し、Seylaz-Tomita法(Tomita et al. J Cereb Blood Flow Metab 25, 858-67, 2005)に従い頭蓋窓を設置した。頭蓋窓設置から2週間以降のマウスを二光子レーザー蛍光顕微鏡下に固定し、スルホローダミン101を5mmol/l含む生理食塩水100μlを腹腔内投与した後、励起波長900nmにて生体二光子蛍光イメージングを行った。その後、C05-05、SPAL-T-83、SPAL-T-95又はSPAL-E-17を0.1%含むDMSO溶液50μlを腹腔内投与し、投与30分後に励起波長900nmにて生体二光子蛍光イメージングを行った。C05-05、SPAL-T-83、SPAL-T-95及びSPAL-E-17に対する検出波長を500~550nm、スルホローダミン101に対する検出波長を573~648nmとした。結果を図4に示す。図4において、矢印は血管、矢頭はαシヌクレイン病変に結合した化合物の蛍光を示している。
[In vivo two-photon laser scanning fluorescence microscopy]
After 4 months of α-synuclein fibril solution injection, model mice were anesthetized with 1.5% (v/v) isoflurane and subjected to the Seylaz-Tomita method (Tomita et al. J Cereb Blood Flow Metab 25, 858-67, 2005). A cranial window was installed. After two weeks from the placement of the cranial window, the mouse was fixed under a two-photon laser fluorescence microscope, and 100 μl of physiological saline containing 5 mmol/l of sulforhodamine 101 was intraperitoneally administered, followed by in vivo two-photon fluorescence imaging at an excitation wavelength of 900 nm. went. After that, 50 μl of DMSO solution containing 0.1% of C05-05, SPAL-T-83, SPAL-T-95 or SPAL-E-17 was intraperitoneally administered, and 30 minutes after administration, biological two-photon irradiation was performed at an excitation wavelength of 900 nm. Fluorescence imaging was performed. The detection wavelength for C05-05, SPAL-T-83, SPAL-T-95 and SPAL-E-17 was 500-550 nm, and the detection wavelength for sulforhodamine 101 was 573-648 nm. The results are shown in FIG. In FIG. 4, arrows indicate blood vessels and arrowheads indicate fluorescence of compounds bound to α-synuclein lesions.
 図4の結果から、C05-05と同じく、SPAL-T-83、SPAL-T-95及びSPAL-E-17を静注すると、これらの化合物が生体の脳内ひいてはニューロン内に移行して、個々のαシヌクレイン病変に結合する様子が生体二光子レーザー蛍光顕微鏡で観察された。したがって、病変の密度が小さいか、又は病変の総数が少なく、病変がPETで検出困難な場合でも、生体蛍光イメージングにより病変を検出可能であると言える。 From the results of FIG. 4, similar to C05-05, when SPAL-T-83, SPAL-T-95 and SPAL-E-17 are intravenously injected, these compounds migrate into the brain and neurons of the living body, Binding to individual α-synuclein lesions was observed by intravital two-photon laser fluorescence microscopy. Therefore, even if the density of lesions is small or the total number of lesions is small, and lesions are difficult to detect by PET, it can be said that lesions can be detected by biofluorescence imaging.
 本発明によれば、αシヌクレイン凝集体への結合選択性が高いαシヌクレイン凝集体結合剤を提供することができる。そして、このαシヌクレイン凝集体結合剤を用いた、イメージング方法を提供することができる。また、αシヌクレイン凝集体結合剤又はその他の用途に用いることができる新規化合物を提供することができる。 According to the present invention, an α-synuclein aggregate-binding agent with high binding selectivity to α-synuclein aggregates can be provided. Then, an imaging method using this α-synuclein aggregate binding agent can be provided. Also, novel compounds that can be used as alpha-synuclein aggregate binding agents or other uses can be provided.

Claims (10)

  1.  下記式(I)、(II)又は(III)で表される化合物、その医薬として許容し得る塩、又はその溶媒和物。
    Figure JPOXMLDOC01-appb-C000001
    A compound represented by the following formula (I), (II) or (III), a pharmaceutically acceptable salt thereof, or a solvate thereof.
    Figure JPOXMLDOC01-appb-C000001
  2.  前記式(I)、(II)又は(III)で表される化合物において1個又はそれ以上の原子が該原子の放射性同位体である、請求項1に記載の化合物、その医薬として許容し得る塩、又はその溶媒和物。 2. The compound of claim 1, wherein one or more atoms in said compound of formula (I), (II) or (III) is a radioactive isotope of said atom, a pharmaceutically acceptable salts or solvates thereof;
  3.  請求項1に記載の化合物、その医薬として許容し得る塩、又はその溶媒和物を含有する、αシヌクレイン凝集体結合剤。 An α-synuclein aggregate binding agent containing the compound according to claim 1, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  4.  請求項2に記載の化合物、その医薬として許容し得る塩、又はその溶媒和物を含有する、αシヌクレイン凝集体結合剤。 An α-synuclein aggregate binding agent containing the compound according to claim 2, a pharmaceutically acceptable salt thereof, or a solvate thereof.
  5.  請求項3に記載のαシヌクレイン凝集体結合剤を含む、αシヌクレイン凝集体の光学イメージング用組成物。 A composition for optical imaging of α-synuclein aggregates, comprising the α-synuclein aggregate-binding agent according to claim 3.
  6.  請求項4に記載のαシヌクレイン凝集体結合剤を含む、αシヌクレイン凝集体の放射イメージング用組成物。 A composition for radiation imaging of α-synuclein aggregates, comprising the α-synuclein aggregate-binding agent according to claim 4.
  7.  請求項3に記載のαシヌクレイン凝集体結合剤を投与された被検体の生体脳に脳外から第1の波長の光を照射した後に、前記脳から発せられる、第1の波長とは異なる第2の波長の光を検出する工程を含む、脳内αシヌクレイン凝集体の光学イメージング方法。 After irradiating the living brain of a subject to whom the α-synuclein aggregate-binding agent according to claim 3 is administered with light of a first wavelength from outside the brain, a second wavelength different from the first wavelength emitted from the brain A method for optical imaging of brain alpha-synuclein aggregates comprising detecting two wavelengths of light.
  8.  請求項4に記載のαシヌクレイン凝集体結合剤を投与された被検体の生体脳から発せられる放射線を検出する工程を含む、脳内αシヌクレイン凝集体の放射イメージング方法。 A radiation imaging method for intracerebral α-synuclein aggregates, comprising the step of detecting radiation emitted from the living brain of a subject administered with the α-synuclein aggregate-binding agent according to claim 4.
  9.  下記式(IV)又は(V)で表される、請求項1又は2に記載の化合物を合成するための、中間体。
    Figure JPOXMLDOC01-appb-C000002
    (式(IV)及び式(V)中、
     Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基又はメタンスルホニル基であり、
     Rは、テトラヒドロ-2H-ピラン-2-イル基又はメトキシメチル基であり、
     Rは、水素原子、tert-ブトキシカルボニル基又は2,4-ジメトキシベンジル基であり、
     式(IV)中、
     Xは窒素原子(N)又は置換されていない炭素原子(CH)であり、
     Rは、水素原子、tert-ブトキシカルボニル基又は2,4-ジメトキシベンジル基である。)
    3. An intermediate for synthesizing the compound according to claim 1 or 2, represented by formula (IV) or (V) below.
    Figure JPOXMLDOC01-appb-C000002
    (In formula (IV) and formula (V),
    R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group,
    R 2 is a tetrahydro-2H-pyran-2-yl group or a methoxymethyl group,
    R 4 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group,
    In formula (IV),
    X is a nitrogen atom (N) or an unsubstituted carbon atom (CH);
    R 3 is a hydrogen atom, a tert-butoxycarbonyl group or a 2,4-dimethoxybenzyl group. )
  10.  下記式(VI)で表される、請求項1又は2に記載の化合物を合成するための、中間体。
    Figure JPOXMLDOC01-appb-C000003
    (式(VI)中、
     Rは、4-ニトロベンゼンスルホニル基、パラトルエンスルホニル基又はメタンスルホニル基であり、
     R4’は、水素原子又はtert-ブトキシカルボニル基であり、
     Xは窒素原子(N)又は置換されていない炭素原子(CH)である。)
    3. An intermediate for synthesizing the compound according to claim 1 or 2, represented by formula (VI) below.
    Figure JPOXMLDOC01-appb-C000003
    (In formula (VI),
    R 1 is a 4-nitrobenzenesulfonyl group, a paratoluenesulfonyl group or a methanesulfonyl group,
    R 4' is a hydrogen atom or a tert-butoxycarbonyl group,
    X is a nitrogen atom (N) or an unsubstituted carbon atom (CH). )
PCT/JP2023/006432 2022-02-24 2023-02-22 NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF WO2023163033A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2022027279 2022-02-24
JP2022-027279 2022-02-24

Publications (1)

Publication Number Publication Date
WO2023163033A1 true WO2023163033A1 (en) 2023-08-31

Family

ID=87766032

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2023/006432 WO2023163033A1 (en) 2022-02-24 2023-02-22 NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF

Country Status (1)

Country Link
WO (1) WO2023163033A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014097474A1 (en) * 2012-12-21 2014-06-26 独立行政法人放射線医学総合研究所 Novel compound for imaging tau protein accumulated in the brain
JP2019512491A (en) * 2016-03-11 2019-05-16 エーシー・イミューン・エス・アー Bicyclic compounds for diagnosis and therapy
WO2020174963A1 (en) * 2019-02-27 2020-09-03 国立研究開発法人量子科学技術研究開発機構 α-SYNUCLEIN AGGREGATE BINDING AGENT AND IMAGING METHOD
WO2022045093A1 (en) * 2020-08-25 2022-03-03 国立研究開発法人量子科学技術研究開発機構 NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014097474A1 (en) * 2012-12-21 2014-06-26 独立行政法人放射線医学総合研究所 Novel compound for imaging tau protein accumulated in the brain
JP2019512491A (en) * 2016-03-11 2019-05-16 エーシー・イミューン・エス・アー Bicyclic compounds for diagnosis and therapy
WO2020174963A1 (en) * 2019-02-27 2020-09-03 国立研究開発法人量子科学技術研究開発機構 α-SYNUCLEIN AGGREGATE BINDING AGENT AND IMAGING METHOD
WO2022045093A1 (en) * 2020-08-25 2022-03-03 国立研究開発法人量子科学技術研究開発機構 NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MIRANDA-AZPIAZU, PATRICIA ET AL.: "Identification and in vitro characterization of C05-01. a PBB3 derivative with improved affinity for alpha-synuclein", BRAIN RESEARCH, vol. 1749, no. 147131, 18 September 2020 (2020-09-18), pages 1 - 9, XP086327336, DOI: 10.1016/j.brainres.2020.147131 *

Similar Documents

Publication Publication Date Title
US11667628B2 (en) Compounds for imaging tau proteins that accumulate in brain
JP5238496B2 (en) Novel amyloid affinity compound
TWI528977B (en) New amyloid affinity products
JP5048655B2 (en) Novel amyloid affinity compound
JPWO2009054496A1 (en) Novel amyloid affinity compound
JP5180838B2 (en) Novel amyloid affinity compound
JP7460176B2 (en) Alpha-synuclein aggregate binding agent and imaging method
JP5954737B2 (en) Radioactive fluorine-labeled quinoxaline compound
KR101854228B1 (en) Novel compound with amyloid affinity
WO2023163033A1 (en) NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF
JPWO2009057576A1 (en) Use and production methods of novel amyloid affinity compounds
WO2022045093A1 (en) NOVEL COMPOUND, α-SYNUCLEIN AGGREGATE BINDER, AND USE THEREOF
JPWO2009057575A1 (en) Use and production methods of novel amyloid affinity compounds
AU2015101580A4 (en) Novel compound for imaging tau protein accumulated in the brain

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 23760032

Country of ref document: EP

Kind code of ref document: A1